Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mol Ther ; 32(10): 3331-3345, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39033321

RESUMEN

Cyclin-dependent kinase-like 5 (CDKL5) deficiency disorder (CDD) is a rare neurodevelopmental disorder caused by a mutation in the X-linked CDKL5 gene. CDKL5 is a serine/threonine kinase that is critical for axon outgrowth and dendritic morphogenesis as well as synapse formation, maturation, and maintenance. This disorder is characterized by early-onset epilepsy, hypotonia, and failure to reach cognitive and motor developmental milestones. Because the disease is monogenic, delivery of the CDKL5 gene to the brain of patients should provide clinical benefit. To this end, we designed a gene therapy vector, adeno-associated virus (AAV)9.Syn.hCDKL5, in which human CDKL5 gene expression is driven by the synapsin promoter. In biodistribution studies conducted in mice, intracerebroventricular (i.c.v.) injection resulted in broader, more optimal biodistribution than did intra-cisterna magna (i.c.m.) delivery. AAV9.Syn.hCDKL5 treatment increased phosphorylation of EB2, a bona fide CDKL5 substrate, demonstrating biological activity in vivo. Our data provide proof of concept that i.c.v. delivery of AAV9.Syn.hCDKL5 to neonatal male Cdkl5 knockout mice reduces pathology and reduces aberrant behavior. Functional improvements were seen at doses of 3e11 to 5e11 vector genomes/g brain, which resulted in transfection of ≥50% of the neurons. Functional improvements were not seen at lower doses, suggesting a requirement for broad distribution for efficacy.


Asunto(s)
Dependovirus , Modelos Animales de Enfermedad , Síndromes Epilépticos , Terapia Genética , Vectores Genéticos , Proteínas Serina-Treonina Quinasas , Espasmos Infantiles , Animales , Terapia Genética/métodos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Ratones , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación , Humanos , Dependovirus/genética , Síndromes Epilépticos/terapia , Síndromes Epilépticos/genética , Espasmos Infantiles/terapia , Espasmos Infantiles/genética , Espasmos Infantiles/metabolismo , Encéfalo/metabolismo , Masculino , Fosforilación , Sinapsinas/genética , Sinapsinas/metabolismo , Discapacidad Intelectual Ligada al Cromosoma X/terapia , Discapacidad Intelectual Ligada al Cromosoma X/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Regiones Promotoras Genéticas , Distribución Tisular , Expresión Génica
2.
Toxicol Pathol ; 49(3): 521-527, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33043844

RESUMEN

Ultrastructural pathology is critical in the morphologic evaluation and characterization of subcellular structures in nonclinical toxicity and efficacy studies. In murine models of ophthalmologic disease, clinical examination is typically paired with other techniques like electroretinography (ERG) and/or optical coherence tomography (OCT) to more fully characterize a finding. High-quality transmission electron microscopy (TEM) can provide a critical, image-based link between these approaches, providing greater confidence in interpretation of ERG or OCT results. In addition to characterization of disease models, TEM can provide detailed visualization of retinal changes identified by clinical examination or light microscopy in nonclinical toxicity studies. The spherical shape of the eye presents unique challenges for trimming, orientation, imaging, and evaluation by TEM. The varied components of the eye require specialized approaches for embedding to facilitate successful sectioning. Controlling for the orientation of the retina is critical to consistent evaluation, driving the need for an improved method of embedding this unique and complex organ. The authors describe a method of sample preparation resulting in optimal orientation of the posterior aspect of murine eyes (rat and mouse) for TEM of the neural retina, Bruch's membrane and/or choroid, with examples from mouse ophthalmic disease models.


Asunto(s)
Electrorretinografía , Retina , Animales , Coroides , Ratones , Microscopía Electrónica de Transmisión , Ratas , Tomografía de Coherencia Óptica
4.
Reprod Sci ; 29(10): 2947-2959, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35641854

RESUMEN

Endometriosis is a condition of the female reproductive tract characterized by endometrium-like tissue growing outside the uterus. Though it is a common cause of pelvic pain and infertility, there is currently no reliable noninvasive method to diagnose the presence of endometriosis without surgery, and the pathophysiological mechanisms that lead to the occurrence of symptoms require further inquiry. Due to patient heterogeneity and delayed diagnosis, animal models are commonly used to study the development of endometriosis, but these are costly due to the large number of animals needed to test various treatments and experimental conditions at multiple endpoints. Here, we describe a method for synthesis of multimodal imaging gold-fluorescein isothiocyanate (FITC) nanoparticles with preclinical application via induction of nanoparticle-labeled endometriosis-like lesions in mice. Labeling donor endometrial tissue fragments with gold-FITC nanoparticles prior to induction of endometriosis in recipients enables in vivo detection of the gold-labeled lesions with photoacoustic imaging. The same imaging method can be used to visualize embryos noninvasively in pregnant mice. Furthermore, the conjugated FITC dye on the gold nanoparticles allows easy isolation of labeled lesion tissue under a fluorescence dissection microscope. After dissection, the presence of gold-FITC nanoparticles and endometrium-like histology of lesions can be verified through fluorescence imaging, gold enhancement, and immunostaining. This method for in vivo imaging of endometriosis-like lesions and fluorescence-guided dissection will permit new experimental possibilities for the longitudinal study of endometriosis development and progression as well as endometriosis-related infertility.


Asunto(s)
Endometriosis , Infertilidad , Nanopartículas del Metal , Técnicas Fotoacústicas , Animales , Modelos Animales de Enfermedad , Endometriosis/patología , Endometrio/patología , Femenino , Fluoresceína-5-Isotiocianato , Oro , Humanos , Estudios Longitudinales , Ratones
5.
Biomedicines ; 9(5)2021 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-33919140

RESUMEN

Genetic, epidemiological and experimental evidence implicate lysosomal dysfunction in Parkinson's disease (PD) and related synucleinopathies. Investigate several mouse models of lysosomal storage diseases (LSDs) and evaluate pathologies reminiscent of synucleinopathies. We obtained brain tissue from symptomatic mouse models of Gaucher, Fabry, Sandhoff, Niemann-Pick A (NPA), Hurler, Pompe and Niemann-Pick C (NPC) diseases and assessed for the presence of Lewy body-like pathology (proteinase K-resistant α-synuclein and tau aggregates) and neuroinflammation (microglial Iba1 and astrocytic GFAP) by immunofluorescence. All seven LSD models exhibited evidence of proteinopathy and/or inflammation in the central nervous system (CNS). However, these phenotypes were divergent. Gaucher and Fabry mouse models displayed proteinase K-resistant α-synuclein and tau aggregates but no neuroinflammation; whereas Sandhoff, NPA and NPC showed marked neuroinflammation and no overt proteinopathy. Pompe disease animals uniquely displayed widespread distribution of tau aggregates accompanied by moderate microglial activation. Hurler mice also demonstrated proteinopathy and microglial activation. The present study demonstrated additional links between LSDs and pathogenic phenotypes that are hallmarks of synucleinopathies. The data suggest that lysosomal dysregulation can contribute to brain region-specific protein aggregation and induce widespread neuroinflammation in the brain. However, only a few LSD models examined exhibited phenotypes consistent with synucleinopathies. While no model can recapitulate the complexity of PD, they can enable the study of specific pathways and mechanisms contributing to disease pathophysiology. The present study provides evidence that there are existing, previously unutilized mouse models that can be employed to study pathogenic mechanisms and gain insights into potential PD subtypes, helping to determine if they are amenable to pathway-specific therapeutic interventions.

6.
Curr Protoc ; 1(4): e118, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33887117

RESUMEN

The ability to analyze the proteome of single cells is critical for the advancement of studies of steady-state and pathological processes. Mass cytometry, or CyTOF, combines principles of mass spectrometry and flow cytometry to enable single-cell analysis of protein expression. CyTOF can simultaneously assess DNA content and proteins and has the capacity to measure 40 to 100 parameters in each cell. Applying this technology to tissues or cells on slides, termed imaging mass cytometry (IMC), allows for visualization of normal and diseased tissues in situ. The high-dimensional proteomic analysis that can be undertaken with CyTOF and IMC has the potential to enhance our understanding of complex and heterogeneous developmental and disease pathways. This article will describe the CyTOF experimental workflow, including reagent selection, sample preparation, and data analysis. CyTOF is compared to flow cytometry, focusing on the strengths and weaknesses of these powerful techniques. Importantly, we review key studies in mouse models of human disease that highlight the strength of CyTOF and IMC to drive discovery research and therapeutic advancement. © 2021 Wiley Periodicals LLC.


Asunto(s)
Citometría de Imagen , Proteómica , Animales , Citometría de Flujo , Ratones , Análisis de la Célula Individual , Tecnología
7.
Front Aging Neurosci ; 12: 524369, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33192449

RESUMEN

Huntington disease (HD) is a fatal, inherited neurodegenerative disorder caused by a mutation in the huntingtin (HTT) gene. While mutant HTT is present ubiquitously throughout life, HD onset typically occurs in mid-life. Oxidative damage accumulates in the aging brain and is a feature of HD. We sought to interrogate the roles and interaction of age and oxidative stress in HD using primary Hu97/18 mouse neurons, neurons differentiated from HD patient induced pluripotent stem cells (iPSCs), and the brains of HD mice. We find that primary neurons must be matured in culture for canonical stress responses to occur. Furthermore, when aging is accelerated in mature HD neurons, mutant HTT accumulates and sensitivity to oxidative stress is selectively enhanced. Furthermore, we observe HD-specific phenotypes in neurons and mouse brains that have undergone accelerated aging, including a selective increase in DNA damage. These findings suggest a role for aging in HD pathogenesis and an interaction between the biological age of HD neurons and sensitivity to exogenous stress.

8.
Physiol Behav ; 219: 112864, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32156555

RESUMEN

Prader-Willi syndrome (PWS) is a rare genetic form of hyperphagia leading to severe obesity, accompanied by endocrine, musculoskeletal, and neurological dysfunction. PWS is caused by the inactivation of contiguous genes on chromosome 15q11-q13, and mice with gene-targeted mutations in one or more of these PWS genes recapitulate PWS-like phenotypes. In addition to evaluating the potential effectiveness of a therapeutic for the treatment of PWS, animal models can be used to elucidate the deficiencies in appetitive and energy balance pathways that lead to hyperphagia and obesity. Various therapeutics have been tested for their effects on ingestive behavior, hyperphagia, and obesity in clinical trials for PWS, with encouraging preliminary results on small groups of participants with PWS. Here, we summarize ingestive behavior-related therapeutics tested in PWS animal models and summarize published data from clinical trials that have evaluated the effect of therapeutics on ingestive behavior in individuals with PWS. We then discuss strategies to accelerate the discovery and translation of therapies into clinical practice in PWS.


Asunto(s)
Síndrome de Prader-Willi , Animales , Ghrelina , Hiperfagia , Ratones , Obesidad , Fenotipo , Síndrome de Prader-Willi/genética
9.
Mol Ther Methods Clin Dev ; 13: 344-358, 2019 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-30989085

RESUMEN

Prader-Willi syndrome (PWS) is a rare neurodevelopmental disorder causing endocrine, musculoskeletal, and neurological dysfunction. PWS is caused by the inactivation of contiguous genes, complicating the development of targeted therapeutics. Clinical trials are now underway in PWS, with more trials to be implemented in the next few years. PWS-like endophenotypes are recapitulated in gene-targeted mice in which the function of one or more PWS genes is disrupted. These animal models can guide priorities for clinical trials or provide information about efficacy of a compound within the context of the specific disease. We now review the current status of preclinical studies that measure the effect of therapeutics on PWS-like endophenotypes. Seven categories of therapeutics (oxytocin and related compounds, K+-ATP channel agonists, melanocortin 4 receptor agonists, incretin mimetics and/or GLP-1 receptor agonists, cannabinoids, ghrelin agents, and Caralluma fimbriata [cactus] extract) have been tested for their effect on endophenotypes in both PWS animal models and clinical trials. Many other therapeutics have been tested in clinical trials, but not preclinical models of PWS or vice versa. Fostering dialogs among investigators performing preclinical validation of animal models and those implementing clinical studies will accelerate the discovery and translation of therapies into clinical practice in PWS.

10.
Heart Rhythm ; 13(9): 1932-40, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27298202

RESUMEN

BACKGROUND: Human ANK2 (ankyrin-B) loss-of-function variants are directly linked with arrhythmia phenotypes. However, in atypical non-ion channel arrhythmia genes such as ANK2 that lack the same degree of robust structure/function and clinical data, it may be more difficult to assign variant disease risk based simply on variant location, minor allele frequency, and/or predictive structural algorithms. The human ankyrin-B p.L1622I variant found in arrhythmia probands displays significant diversity in minor allele frequency across populations. OBJECTIVE: The objective of this study was to directly test the in vivo impact of ankyrin-B p.L1622I on cardiac electrical phenotypes and arrhythmia risk using a new animal model. METHODS: We tested arrhythmia phenotypes in a new "knock-in" animal model harboring the human ankyrin-B p.L1622I variant. RESULTS: Ankyrin-B p.L1622I displays reduced posttranslational expression in vivo, resulting in reduced cardiac ankyrin-B expression and reduced association with binding-partner Na/Ca exchanger. Ankyrin-B(L1622I/L1622I) mice display changes in heart rate, atrioventricular and intraventricular conduction, and alterations in repolarization. Furthermore, ankyrin-B(L1622I/L1622I) mice display catecholamine-dependent arrhythmias. At the cellular level, ankyrin-B(L1622I/L1622I) myocytes display increased action potential duration and severe arrhythmogenic afterdepolarizations that provide a mechanistic rationale for the arrhythmias. CONCLUSION: Our findings support in vivo arrhythmogenic phenotypes of an ANK2 variant with unusual frequency in select populations. On the basis of our findings and current clinical data, we support classification of p.L1622I as a "mild" loss-of-function variant that may confer arrhythmia susceptibility in the context of secondary risk factors including environment, medication, and/or additional genetic variation.


Asunto(s)
Ancirinas/genética , Arritmias Cardíacas/genética , Potenciales de Acción/genética , Animales , Arritmias Cardíacas/etnología , Arritmias Cardíacas/fisiopatología , Población Negra/genética , Modelos Animales de Enfermedad , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad/etnología , Variación Genética , Humanos , Mutación con Pérdida de Función , Ratones , Ratones Endogámicos C57BL , Fenotipo , Medición de Riesgo/etnología , Factores de Riesgo
11.
F1000Res ; 5: 2884, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28491277

RESUMEN

Background: The proliferation of publicly accessible large-scale biological data together with increasing availability of bioinformatics tools have the potential to transform biomedical research. Here we report a crowdsourcing Jamboree that explored whether a team of volunteer biologists without formal bioinformatics training could use OMiCC, a crowdsourcing web platform that facilitates the reuse and (meta-) analysis of public gene expression data, to compile and annotate gene expression data, and design comparisons between disease and control sample groups. Methods: The Jamboree focused on several common human autoimmune diseases, including systemic lupus erythematosus (SLE), multiple sclerosis (MS), type I diabetes (DM1), and rheumatoid arthritis (RA), and the corresponding mouse models. Meta-analyses were performed in OMiCC using comparisons constructed by the participants to identify 1) gene expression signatures for each disease (disease versus healthy controls at the gene expression and biological pathway levels), 2) conserved signatures across all diseases within each species (pan-disease signatures), and 3) conserved signatures between species for each disease and across all diseases (cross-species signatures). Results: A large number of differentially expressed genes were identified for each disease based on meta-analysis, with observed overlap among diseases both within and across species. Gene set/pathway enrichment of upregulated genes suggested conserved signatures (e.g., interferon) across all human and mouse conditions. Conclusions: Our Jamboree exercise provides evidence that when enabled by appropriate tools, a "crowd" of biologists can work together to accelerate the pace by which the increasingly large amounts of public data can be reused and meta-analyzed for generating and testing hypotheses. Our encouraging experience suggests that a similar crowdsourcing approach can be used to explore other biological questions.

12.
Hypertension ; 62(1): 140-6, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23670301

RESUMEN

Monoamine oxidases (MAOs) generate H(2)O(2) as a by-product of their catalytic cycle. Whether MAOs are mediators of endothelial dysfunction is unknown and was determined here in the angiotensin II and lipopolysaccharide-models of vascular dysfunction in mice. Quantitative real-time polymerase chain reaction revealed that mouse aortas contain enzymes involved in catecholamine generation and MAO-A and MAO-B mRNA. MAO-A and -B proteins could be detected by Western blot not only in mouse aortas but also in human umbilical vein endothelial cells. Ex vivo incubation of mouse aorta with recombinant MAO-A increased H(2)O(2) formation and induced endothelial dysfunction that was attenuated by polyethylene glycol-catalase and MAO inhibitors. In vivo lipopolysaccharide (8 mg/kg IP overnight) or angiotensin II (1 mg/kg per day, 2 weeks, minipump) treatment induced vascular MAO-A and -B expressions and resulted in attenuated endothelium-dependent relaxation of the aorta in response to acetylcholine. MAO inhibitors reduced the lipopolysaccharide- and angiotensin II-induced aortic reactive oxygen species formation by 50% (ferrous oxidation xylenol orange assay) and partially normalized endothelium-dependent relaxation. MAO-A and MAO-B inhibitors had an additive effect; combined application completely restored endothelium-dependent relaxation. To determine how MAO-dependent H(2)O(2) formation induces endothelial dysfunction, cyclic GMP was measured. Histamine stimulation of human umbilical vein endothelial cells to activate endothelial NO synthase resulted in an increase in cyclic GMP, which was almost abrogated by MAO-A exposure. MAO inhibition prevented this effect, suggesting that MAO-induced H(2)O(2) formation is sufficient to attenuate endothelial NO release. Thus, MAO-A and MAO-B are both expressed in the mouse aorta, induced by in vivo lipopolysaccharide and angiotensin II treatment and contribute via the generation of H(2)O(2) to endothelial dysfunction in vascular disease models.


Asunto(s)
Endotelio Vascular/fisiopatología , Regulación de la Expresión Génica , Monoaminooxidasa/genética , Estrés Oxidativo/genética , ARN Mensajero/genética , Enfermedades Vasculares/genética , Vasodilatación/fisiología , Angiotensina II/farmacología , Animales , Aorta Torácica/enzimología , Aorta Torácica/patología , Aorta Torácica/fisiopatología , Western Blotting , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Humanos , Inmunohistoquímica , Ratones , Monoaminooxidasa/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Venas Umbilicales/enzimología , Venas Umbilicales/patología , Venas Umbilicales/fisiopatología , Enfermedades Vasculares/enzimología , Enfermedades Vasculares/fisiopatología , Vasodilatación/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda