Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Neuron ; 111(1): 49-64.e5, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36351424

ABSTRACT

In albinism, aberrations in the ipsi-/contralateral retinal ganglion cell (RGC) ratio compromise the functional integrity of the binocular circuit. Here, we focus on the mouse ciliary margin zone (CMZ), a neurogenic niche at the embryonic peripheral retina, to investigate developmental processes regulating RGC neurogenesis and identity acquisition. We found that the mouse ventral CMZ generates predominantly ipsilaterally projecting RGCs, but this output is altered in the albino visual system because of CyclinD2 downregulation and disturbed timing of the cell cycle. Consequently, albino as well as CyclinD2-deficient pigmented mice exhibit diminished ipsilateral retinogeniculate projection and poor depth perception. In albino mice, pharmacological stimulation of calcium channels, known to upregulate CyclinD2 in other cell types, augmented CyclinD2-dependent neurogenesis of ipsilateral RGCs and improved stereopsis. Together, these results implicate CMZ neurogenesis and its regulators as critical for the formation and function of the mammalian binocular circuit.


Subject(s)
Albinism , Retina , Animals , Mice , Albinism/metabolism , Cell Division , Mammals , Neurogenesis/physiology , Retina/metabolism , Retinal Ganglion Cells/metabolism , Visual Pathways
2.
J Vis Exp ; (185)2022 07 25.
Article in English | MEDLINE | ID: mdl-35938825

ABSTRACT

The family of caspases is known to mediate many cellular pathways beyond cell death, including cell differentiation, axonal pathfinding, and proliferation. Since the identification of the family of cell death proteases, there has been a search for tools to identify and expand the function of specific family members in development, health, and disease states. However, many of the currently commercially available caspase tools that are widely used are not specific for the targeted caspase. In this report, we delineate the approach we have used to identify, validate, and target caspase-9 in the nervous system using a novel inhibitor and genetic approaches with immunohistochemical read-outs. Specifically, we used the retinal neuronal tissue as a model to identify and validate the presence and function of caspases. This approach enables the interrogation of cell-type specific apoptotic and non-apoptotic caspase-9 functions and can be applied to other complex tissues and caspases of interest. Understanding the functions of caspases can help to expand current knowledge in cell biology, and can also be advantageous to identify potential therapeutic targets due to their involvement in disease.


Subject(s)
Caspases , Retina , Apoptosis , Caspase 3/metabolism , Caspase 9/metabolism , Caspases/metabolism , Cell Differentiation , Nervous System , Retina/metabolism
3.
J Vis Exp ; (182)2022 04 21.
Article in English | MEDLINE | ID: mdl-35532239

ABSTRACT

Advancements in ophthalmic imaging tools offer an unprecedented level of access to researchers working with animal models of neurovascular injury. To properly leverage this greater translatability, there is a need to devise reproducible methods of drawing quantitative data from these images. Optical coherence tomography (OCT) imaging can resolve retinal histology at micrometer resolution and reveal functional differences in vascular blood flow. Here, we delineate noninvasive vascular readouts that we use to characterize pathological damage post vascular insult in an optimized mouse model of retinal vein occlusion (RVO). These readouts include live imaging analysis of retinal morphology, disorganization of retinal inner layers (DRIL) measure of capillary ischemia, and fluorescein angiography measures of retinal edema and vascular density. These techniques correspond directly to those used to examine patients with retinal disease in the clinic. Standardizing these methods enables direct and reproducible comparison of animal models with clinical phenotypes of ophthalmic disease, increasing the translational power of vascular injury models.


Subject(s)
Vascular System Injuries , Animals , Humans , Mice , Reproducibility of Results , Retina/diagnostic imaging , Retina/pathology , Retinal Vessels/diagnostic imaging , Retrospective Studies , Tomography, Optical Coherence/methods , Vascular System Injuries/pathology , Visual Acuity
4.
J Vis Exp ; (174)2021 08 06.
Article in English | MEDLINE | ID: mdl-34424250

ABSTRACT

Mouse models of retinal vein occlusion (RVO) are often used in ophthalmology to study hypoxic-ischemic injury in the neural retina. In this report, a detailed method pointing out critical steps is provided with recommendations for optimization to achieve consistently successful occlusion rates across different genetically modified mouse strains. The RVO mouse model consists primarily of the intravenous administration of a photosensitizer dye followed by laser photocoagulation using a retinal imaging microscope attached to an ophthalmic guided laser. Three variables were identified as determinants of occlusion consistency. By adjusting the wait time after rose bengal administration and balancing the baseline and experimental laser output, the variability across experiments can be limited and a higher success rate of occlusions achieved. This method can be used to study retinal diseases that are characterized by retinal edema and hypoxic-ischemic injury. Additionally, as this model induces vascular injury, it can also be applied to study the neurovasculature, neuronal death, and inflammation.


Subject(s)
Retinal Vein Occlusion , Animals , Disease Models, Animal , Hypoxia , Mice , Retina , Rose Bengal
6.
Nat Commun ; 11(1): 3173, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32576823

ABSTRACT

Central nervous system ischemic injury features neuronal dysfunction, inflammation and breakdown of vascular integrity. Here we show that activation of endothelial caspase-9 after hypoxia-ischemia is a critical event in subsequent dysfunction of the blood-retina barrier, using a panel of interrelated ophthalmic in vivo imaging measures in a mouse model of retinal vein occlusion (RVO). Rapid nonapoptotic activation of caspase-9 and its downstream effector caspase-7 in endothelial cells promotes capillary ischemia and retinal neurodegeneration. Topical eye-drop delivery of a highly selective caspase-9 inhibitor provides morphological and functional retinal protection. Inducible endothelial-specific caspase-9 deletion phenocopies this protection, with attenuated retinal edema, reduced inflammation and preserved neuroretinal morphology and function following RVO. These results reveal a non-apoptotic function of endothelial caspase-9 which regulates blood-retina barrier integrity and neuronal survival, and identify caspase-9 as a therapeutic target in neurovascular disease.


Subject(s)
Caspase 9/metabolism , Hypoxia/metabolism , Ischemia/metabolism , Retinal Vein Occlusion/metabolism , Vascular System Injuries/metabolism , Animals , Blood-Retinal Barrier/metabolism , Caspase 7/metabolism , Caspase 9/drug effects , Caspase 9/genetics , Cell Death , Disease Models, Animal , Endothelial Cells/metabolism , Female , Genetic Predisposition to Disease/genetics , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rabbits , Retina/metabolism , Retina/pathology , Retinal Vein Occlusion/drug therapy , Retinal Vein Occlusion/pathology , Vascular System Injuries/pathology
7.
Vaccine ; 36(9): 1146-1153, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29395518

ABSTRACT

Vaccination remains the best available prophylaxis to prevent influenza virus infections, yet current inadequacies in influenza virus vaccine manufacturing often lead to vaccine shortages at times when the vaccine is most needed, as it was the case during the last influenza virus pandemic. Novel influenza virus vaccine production systems will be crucial to improve public health and safety. Here we report the optimization of influenza B virus growth in the proprietary EB66® cell line, currently in use for human vaccine production. To this end, we collected, curated and sequenced 71 influenza B viruses selected for high diversity in date of isolation and lineage. This viral collection was tested for ability to enter and replicate within EB66® cells in a single cycle assay and appears to readily infect these cells. When the collection was tested for viral progeny production in a multi-cycle assay, we found a large variation from strain to strain. The strains with the top growth characteristics from the B/Victoria and B/Yamagata lineages were selected for vaccine backbone generation using a reverse genetics system. We then showed that these backbones maintain their desirable growth within EB66® cells when the HA and NA from poorly growing strains were substituted for the parental segments, indicating that the selected backbones are viable options for vaccine production in EB66®. Finally, we show that compounds previously reported to enhance influenza virus growth in cell culture also increase virus production in the EB66® cell line.


Subject(s)
Influenza B virus/growth & development , Influenza B virus/genetics , Reverse Genetics , Animals , Cell Line , Ducks , Hemagglutination, Viral , Influenza Vaccines
SELECTION OF CITATIONS
SEARCH DETAIL
...