Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Clin Ophthalmol ; 16: 1401-1411, 2022.
Article in English | MEDLINE | ID: mdl-35535124

ABSTRACT

Purpose: Ocular trauma with intraocular foreign body (IOFB) can have devastating visual consequences. Management and antimicrobial strategies remain variable due to the infrequency and heterogeneity of presentation. Our goal was to identify risk factors for endophthalmitis and poor visual outcomes in cases of IOFB and investigate management strategies. Patients and Methods: A retrospective chart review was conducted in 88 eyes of 88 patients suffering traumatic injury with IOFB at the University of Michigan between January 2000 and December 2019. Medical records were reviewed to characterize the injuries and IOFBs as well as how clinical presentation and treatment modalities were associated with outcomes. Results: Delayed presentation (P=0.016) and organic IOFB (P=0.044) were associated with development of endophthalmitis. Retinal detachment (P=0.012), wound length greater than 5 mm (P=0.041), and poor presenting visual acuity (P=0.003) correlated with poor final visual outcome. Antibiotic prophylaxis was given to all patients, though agents and routes of delivery varied. Endophthalmitis developed in 4.9% of the eyes after initial management, with primary and secondary removal of posterior segment IOFBs associated with similar rates of endophthalmitis (P=1.000). Conclusion: Poor presenting visual acuity and severity of injury, as measured by large wound and retinal detachment, correlate with poor visual outcome. Prompt globe closure and antimicrobial prophylaxis are critical for infection prevention. In cases where IOFB removal and globe closure cannot be performed concurrently, primary globe closure with aggressive antibiotic prophylaxis offers a reasonable alternative to prevent endophthalmitis.

2.
Clin Ophthalmol ; 15: 2077-2087, 2021.
Article in English | MEDLINE | ID: mdl-34040343

ABSTRACT

BACKGROUND/AIMS: To determine the rate of endophthalmitis and assess risk factors for development of endophthalmitis following open globe injury (OGI). METHODS: A retrospective chart review of all patients treated for OGI at the University of Michigan from January 2000 to July 2017 was conducted. Exclusion criteria included intravitreal injection or intraocular surgery in the 30 days prior to injury or less than 30 days of follow-up. A total of 586 out of 993 open globe injuries were included in the study. The main outcome measure was the rate of endophthalmitis. RESULTS: In this study, 25/586 eyes (4.3%) had endophthalmitis. Of these, 12/25 eyes (48.0%) presented with endophthalmitis and 13/25 eyes (52.0%) developed endophthalmitis after globe closure. Multivariate analysis identified time to globe repair (OR 4.5, CI 1.9-10.7, p = 0.0008), zone I injury (OR 3.6, CI 1.1-11.0, p = 0.0282), and need for additional surgery (OR 5.5, CI 1.5-19.7, p = 0.0092) as factors associated with increased risk of developing endophthalmitis. Subconjunctival antibiotic injection at the time of globe closure (OR 0.3, CI 0.1-0.7, p = 0.0036) was associated with decreased risk of developing endophthalmitis. CONCLUSION: Prompt globe closure and subconjunctival antibiotics may reduce the risk of endophthalmitis in OGI. Furthermore, our practice of a one-time dose of systemic prophylactic antibiotics, and intravitreal antibiotics if intraocular foreign body (IOFB) removal is delayed, was not found to increase the rate of endophthalmitis.

3.
Diabetes ; 70(7): 1549-1560, 2021 07.
Article in English | MEDLINE | ID: mdl-33883214

ABSTRACT

Diabetic retinopathy is one of the leading causes of vision loss and blindness. Extensive preclinical and clinical evidence exists for both vascular and neuronal pathology. However, the relationship of these changes in the neurovascular unit and impact on vision remains to be determined. Here, we investigate the role of tight junction protein occludin phosphorylation at S490 in modulating barrier properties and its impact on visual function. Conditional vascular expression of the phosphorylation-resistant Ser490 to Ala (S490A) form of occludin preserved tight junction organization and reduced vascular endothelial growth factor (VEGF)-induced permeability and edema formation after intraocular injection. In the retinas of streptozotocin-induced diabetic mice, endothelial-specific expression of the S490A form of occludin completely prevented diabetes-induced permeability to labeled dextran and inhibited leukostasis. Importantly, vascular-specific expression of the occludin mutant completely blocked the diabetes-induced decrease in visual acuity and contrast sensitivity. Together, these results reveal that occludin acts to regulate barrier properties downstream of VEGF in a phosphorylation-dependent manner and that loss of inner blood-retinal barrier integrity induced by diabetes contributes to vision loss.


Subject(s)
Blood-Retinal Barrier/physiology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Retinopathy/physiopathology , Occludin/physiology , Visual Acuity , Animals , Leukostasis/prevention & control , Mice , Mice, Inbred C57BL , Permeability , Phosphorylation , Streptozocin , Vascular Endothelial Growth Factor A/physiology
4.
Nat Commun ; 11(1): 3839, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32737294

ABSTRACT

Chromatin regulates spatiotemporal gene expression during neurodevelopment, but it also mediates DNA damage repair essential to proliferating neural progenitor cells (NPCs). Here, we uncover molecularly dissociable roles for nucleosome remodeler Ino80 in chromatin-mediated transcriptional regulation and genome maintenance in corticogenesis. We find that conditional Ino80 deletion from cortical NPCs impairs DNA double-strand break (DSB) repair, triggering p53-dependent apoptosis and microcephaly. Using an in vivo DSB repair pathway assay, we find that Ino80 is selectively required for homologous recombination (HR) DNA repair, which is mechanistically distinct from Ino80 function in YY1-associated transcription. Unexpectedly, sensitivity to loss of Ino80-mediated HR is dependent on NPC division mode: Ino80 deletion leads to unrepaired DNA breaks and apoptosis in symmetric NPC-NPC divisions, but not in asymmetric neurogenic divisions. This division mode dependence is phenocopied following conditional deletion of HR gene Brca2. Thus, distinct modes of NPC division have divergent requirements for Ino80-dependent HR DNA repair.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , BRCA2 Protein/genetics , Chromatin/chemistry , DNA-Binding Proteins/genetics , Neural Stem Cells/metabolism , Neurogenesis/genetics , Recombinational DNA Repair , ATPases Associated with Diverse Cellular Activities/deficiency , Animals , Apoptosis/genetics , BRCA2 Protein/deficiency , Cell Division , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA/genetics , DNA/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins/deficiency , Embryo, Mammalian , Gene Expression Regulation, Developmental , Mice , Mice, Transgenic , Neocortex/cytology , Neocortex/growth & development , Neocortex/metabolism , Neural Stem Cells/cytology , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
5.
J Neurosci ; 40(19): 3720-3740, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32273484

ABSTRACT

Nestin, an intermediate filament protein widely used as a marker of neural progenitors, was recently found to be expressed transiently in developing cortical neurons in culture and in developing mouse cortex. In young cortical cultures, nestin regulates axonal growth cone morphology. In addition, nestin, which is known to bind the neuronal cdk5/p35 kinase, affects responses to axon guidance cues upstream of cdk5, specifically, to Sema3a. Changes in growth cone morphology require rearrangements of cytoskeletal networks, and changes in microtubules and actin filaments are well studied. In contrast, the roles of intermediate filament proteins in this process are poorly understood, even in cultured neurons. Here, we investigate the molecular mechanism by which nestin affects growth cone morphology and Sema3a sensitivity. We find that nestin selectively facilitates the phosphorylation of the lissencephaly-linked protein doublecortin (DCX) by cdk5/p35, but the phosphorylation of other cdk5 substrates is not affected by nestin. We uncover that this substrate selectivity is based on the ability of nestin to interact with DCX, but not with other cdk5 substrates. Nestin thus creates a selective scaffold for DCX with activated cdk5/p35. Last, we use cortical cultures derived from Dcx KO mice to show that the effects of nestin on growth cone morphology and on Sema3a sensitivity are DCX-dependent, thus suggesting a functional role for the DCX-nestin complex in neurons. We propose that nestin changes growth cone behavior by regulating the intracellular kinase signaling environment in developing neurons. The sex of animal subjects is unknown.SIGNIFICANCE STATEMENT Nestin, an intermediate filament protein highly expressed in neural progenitors, was recently identified in developing neurons where it regulates growth cone morphology and responsiveness to the guidance cue Sema3a. Changes in growth cone morphology require rearrangements of cytoskeletal networks, but the roles of intermediate filaments in this process are poorly understood. We now report that nestin selectively facilitates phosphorylation of the lissencephaly-linked doublecortin (DCX) by cdk5/p35, but the phosphorylation of other cdk5 substrates is not affected. This substrate selectivity is based on preferential scaffolding of DCX, cdk5, and p35 by nestin. Additionally, we demonstrate a functional role for the DCX-nestin complex in neurons. We propose that nestin changes growth cone behavior by regulating intracellular kinase signaling in developing neurons.


Subject(s)
Microtubule-Associated Proteins/metabolism , Nestin/metabolism , Neurogenesis/physiology , Neurons/metabolism , Neuropeptides/metabolism , Animals , COS Cells , Chlorocebus aethiops , Doublecortin Domain Proteins , Doublecortin Protein , Female , Growth Cones/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Phosphorylation , Semaphorin-3A/metabolism
6.
Nat Commun ; 10(1): 2588, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31197172

ABSTRACT

The brain is a genomic mosaic shaped by cellular responses to genome damage. Here, we manipulate somatic genome stability by conditional Knl1 deletion from embryonic mouse brain. KNL1 mutations cause microcephaly and KNL1 mediates the spindle assembly checkpoint, a safeguard against chromosome missegregation and aneuploidy. We find that following Knl1 deletion, segregation errors in mitotic neural progenitor cells give rise to DNA damage on the missegregated chromosomes. This triggers rapid p53 activation and robust apoptotic and microglial phagocytic responses that extensively eliminate cells with somatic genome damage, thus causing microcephaly. By leaving only karyotypically normal progenitors to continue dividing, these mechanisms provide a second safeguard against brain somatic aneuploidy. Without Knl1 or p53-dependent safeguards, genome-damaged cells are not cleared, alleviating microcephaly, but paradoxically leading to total pre-weaning lethality. Thus, mitotic genome damage activates robust responses to eliminate somatic mutant cells, which if left unpurged, can impact brain and organismal fitness.


Subject(s)
Aneuploidy , Microcephaly/genetics , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/genetics , Chromosome Segregation/genetics , DNA Damage/genetics , Disease Models, Animal , Embryo, Mammalian , Genomic Instability , Humans , Kinetochores/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Primary Cell Culture , Sequence Deletion , Spindle Apparatus/metabolism
7.
Am J Pathol ; 188(10): 2392-2405, 2018 10.
Article in English | MEDLINE | ID: mdl-30220554

ABSTRACT

Changes in permeability of retinal blood vessels contribute to macular edema and the pathophysiology of numerous ocular diseases, including diabetic retinopathy, retinal vein occlusions, and macular degeneration. Vascular endothelial growth factor (VEGF) induces retinal permeability and macular thickening in these diseases. However, inflammatory agents, such as tumor necrosis factor-α (TNF-α), also may drive vascular permeability, specifically in patients unresponsive to anti-VEGF therapy. Recent evidence suggests VEGF and TNF-α induce permeability through distinct mechanisms; however, both require the activation of atypical protein kinase C (aPKC). We provide evidence, using genetic mouse models and therapeutic intervention with small molecules, that inhibition of aPKC prevented or reduced vascular permeability in animal models of retinal inflammation. Expression of a kinase-dead aPKC transgene, driven by a vascular and hematopoietic restricted promoter, reduced retinal vascular permeability in an ischemia-reperfusion model of retinal injury. This effect was recapitulated with a small-molecule inhibitor of aPKC. Expression of the kinase-dead aPKC transgene dramatically reduced the expression of inflammatory factors and blocked the attraction of inflammatory monocytes and granulocytes after ischemic injury. Coinjection of VEGF with TNF-α was sufficient to induce permeability, edema, and retinal inflammation, and treatment with an aPKC inhibitor prevented VEGF/TNF-α-induced permeability. These data suggest that aPKC contributes to inflammation-driven retinal vascular pathology and may be an attractive target for therapeutic intervention.


Subject(s)
Capillary Permeability/physiology , Protein Kinase C/antagonists & inhibitors , Retinal Vessels/physiology , Animals , Capillary Permeability/drug effects , Male , Mice, Inbred C57BL , Papilledema/chemically induced , Papilledema/physiopathology , Rats, Long-Evans , Recombinant Proteins , Reperfusion Injury/physiopathology , Retinitis/chemically induced , Retinitis/physiopathology , Tight Junctions/chemistry , Tight Junctions/physiology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
8.
J Neurosci ; 38(10): 2399-2412, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29437890

ABSTRACT

Transcriptional programs instruct the generation and maintenance of diverse subtypes of neural cells, establishment of distinct brain regions, formation and function of neural circuits, and ultimately behavior. Spatiotemporal and cell type-specific analyses of the transcriptome, the sum total of all RNA transcripts in a cell or an organ, can provide insights into the role of genes in brain development and function, and their potential contribution to disorders of the brain. In the previous decade, advances in sequencing technology and funding from the National Institutes of Health and private foundations for large-scale genomics projects have led to a growing collection of brain transcriptome databases. These valuable resources provide rich and high-quality datasets with spatiotemporal, cell type-specific, and single-cell precision. Most importantly, many of these databases are publicly available via user-friendly web interface, making the information accessible to individual scientists without the need for advanced computational expertise. Here, we highlight key publicly available brain transcriptome databases, summarize the tissue sources and methods used to generate the data, and discuss their utility for neuroscience research.


Subject(s)
Brain Chemistry/genetics , Databases, Genetic , Transcriptome/genetics , Animals , Computational Biology , Gene Expression Regulation , Humans
9.
Invest Ophthalmol Vis Sci ; 54(6): 4007-15, 2013 Jun 12.
Article in English | MEDLINE | ID: mdl-23640037

ABSTRACT

PURPOSE: Glucocorticoids (GCs) effectively reduce retinal edema and induce vascular barrier properties but possess unwanted side effects. Understanding GC induction of barrier properties may lead to more effective and specific therapies. Previous work identified the occludin enhancer element (OEE) as a GC-responsive cis-element in the promoters of multiple junctional genes, including occludin, claudin-5, and cadherin-9. Here, we identify two OEE-binding factors and determine their contribution to GC induction of tight junction (TJ) gene expression and endothelial barrier properties. METHODS: OEE-binding factors were isolated from human retinal endothelial cells (HREC) using DNA affinity purification followed by MALDI-TOF MS/MS. Chromatin immunoprecipitation (ChIP) assays determined in situ binding. siRNA was used to evaluate the role of trans-acting factors in transcription of TJ genes in response to GC stimulation. Paracellular permeability was determined by quantifying flux through a cell monolayer, whereas transendothelial electrical resistance (TER) was measured using the ECIS system. RESULTS: MS/MS analysis of HREC nuclear extracts identified the heterodimer of transcription factors p54/NONO (p54) and polypyrimidine tract-binding protein-associated splicing factor (PSF) as OEE-binding factors, which was confirmed by ChIP assay from GC-treated endothelial cells and rat retina. siRNA knockdown of p54 demonstrated that this factor is necessary for GC induction of occludin and claudin-5 expression. Further, p54 knockdown ablated the pro-barrier effects of GC treatment. CONCLUSIONS: p54 is essential for GC-mediated expression of occludin, claudin-5, and barrier induction, and the p54/PSF heterodimer may contribute to normal blood-retinal barrier (BRB) induction in vivo. Understanding the mechanism of GC induction of BRB properties may provide novel therapies for macular edema.


Subject(s)
Blood-Retinal Barrier/drug effects , Dexamethasone/pharmacology , Endothelium, Vascular/drug effects , Glucocorticoids/pharmacology , Nuclear Matrix-Associated Proteins/metabolism , Occludin/biosynthesis , Octamer Transcription Factors/metabolism , RNA-Binding Proteins/metabolism , Animals , Blood-Retinal Barrier/metabolism , Blotting, Western , Cattle , Cells, Cultured , Chromatin Immunoprecipitation , Claudin-5/metabolism , DNA-Binding Proteins , Electrophoretic Mobility Shift Assay , Endothelium, Vascular/metabolism , Gene Silencing/physiology , Humans , Male , Nuclear Matrix-Associated Proteins/genetics , Octamer Transcription Factors/genetics , PTB-Associated Splicing Factor , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Rats , Retinal Vessels/cytology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Tight Junctions/drug effects , Trans-Activators , Transfection
10.
Proc Natl Acad Sci U S A ; 109(27): 10855-60, 2012 Jul 03.
Article in English | MEDLINE | ID: mdl-22711802

ABSTRACT

Tight junctions (TJs) are dynamic cellular structures that are critical for compartmentalizing environments within tissues and regulating transport of small molecules, ions, and fluids. Phosphorylation-dependent binding of the transmembrane protein occludin to the structural organizing protein ZO-1 contributes to the regulation of barrier properties; however, the details of their interaction are controversial. Using small angle X-ray scattering (SAXS), NMR chemical shift perturbation, cross-saturation, in vitro binding, and site-directed mutagenesis experiments. we define the interface between the ZO-1 PDZ3-SH3-U5-GuK (PSG) and occludin coiled-coil (CC) domains. The interface is comprised of basic residues in PSG and an acidic region in CC. Complex formation is blocked by a peptide (REESEEYM) that corresponds to CC residues 468-475 and includes a previously uncharacterized phosphosite, with the phosphorylated version having a larger effect. Furthermore, mutation of E470 and E472 reduces cell border localization of occludin. Together, these results localize the interaction to an acidic region in CC and a predominantly basic helix V within the ZO-1 GuK domain. This model has important implications for the phosphorylation-dependent regulation of the occludin:ZO-1 complex.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Tight Junctions/metabolism , Acids/chemistry , Calmodulin/metabolism , Cell Membrane Permeability/physiology , Escherichia coli/genetics , Guanylate Kinases/metabolism , Humans , MARVEL Domain Containing 2 Protein , Membrane Proteins/genetics , Mutagenesis/physiology , Nuclear Magnetic Resonance, Biomolecular , Occludin , Phosphoproteins/genetics , Phosphorylation/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , Scattering, Radiation , Solutions/chemistry , Zonula Occludens-1 Protein
11.
Biochem J ; 446(3): 455-67, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22721706

ABSTRACT

Pro-inflammatory cytokines and growth factors such as VEGF (vascular endothelial growth factor) contribute to the loss of the BRB (blood-retinal barrier) and subsequent macular oedema in various retinal pathologies. VEGF signalling requires PKCß [conventional PKC (protein kinase C)] activity; however, PKCß inhibition only partially prevents VEGF-induced endothelial permeability and does not affect pro-inflammatory cytokine-induced permeability, suggesting the involvement of alternative signalling pathways. In the present study, we provide evidence for the involvement of aPKC (atypical PKC) signalling in VEGF-induced endothelial permeability and identify a novel class of inhibitors of aPKC that prevent BRB breakdown in vivo. Genetic and pharmacological manipulations of aPKC isoforms were used to assess their contribution to endothelial permeability in culture. A chemical library was screened using an in vitro kinase assay to identify novel small-molecule inhibitors, and further medicinal chemistry was performed to delineate a novel pharmacophore. We demonstrate that aPKC isoforms are both sufficient and required for VEGF-induced endothelial permeability. Furthermore, these specific, potent, non-competitive, small-molecule inhibitors prevented VEGF-induced tight junction internalization and retinal endothelial permeability in response to VEGF in both primary culture and in rodent retina. The results of the present study suggest that aPKC inhibition with 2-amino-4-phenyl-thiophene derivatives may be developed to preserve the BRB in retinal diseases such as diabetic retinopathy or uveitis, and the BBB (blood-brain barrier) in the presence of brain tumours.


Subject(s)
Blood-Retinal Barrier/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cattle , Diabetic Retinopathy/metabolism , Humans , Male , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Protein Kinase C/metabolism , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retina/cytology , Retina/metabolism , Retinal Vessels/cytology , Retinal Vessels/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...