Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Publication year range
1.
Development ; 146(23)2019 12 02.
Article in English | MEDLINE | ID: mdl-31740533

ABSTRACT

To maintain body homeostasis, endocrine systems must detect and integrate blood-borne peripheral signals. This is mediated by fenestrae, specialized permeable pores in the endothelial membrane. Plasmalemma vesicle-associated protein (Plvap) is located in the fenestral diaphragm and is thought to play a role in the passage of proteins through the fenestrae. However, this suggested function has yet to be demonstrated directly. We studied the development of fenestrated capillaries in the hypophysis, a major neuroendocrine interface between the blood and brain. Using a transgenic biosensor to visualize the vascular excretion of the genetically tagged plasma protein DBP-EGFP, we show that the developmental acquisition of vascular permeability coincides with differential expression of zebrafish plvap orthologs in the hypophysis versus brain. Ultrastructural analysis revealed that plvapb mutants display deficiencies in fenestral diaphragms and increased density of hypophyseal fenestrae. Measurements of DBP-EGFP extravasation in plvapb mutants provided direct proof that Plvap limits the rate of blood-borne protein passage through fenestrated endothelia. We present the regulatory role of Plvap in the development of blood-borne protein detection machinery at a neuroendocrine interface through which hormones are released to the general circulation.


Subject(s)
Capillary Permeability/physiology , Endothelium, Vascular/metabolism , Membrane Proteins/metabolism , Pituitary Gland/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Membrane Proteins/genetics , Mutation , Protein Transport/physiology , Zebrafish/genetics
2.
Exp Eye Res ; 215: 108899, 2022 02.
Article in English | MEDLINE | ID: mdl-34929159

ABSTRACT

Sorsby Fundus Dystrophy (SFD) is a rare form of macular degeneration that is clinically similar to age-related macular degeneration (AMD), and a histologic hallmark of SFD is a thick layer of extracellular deposits beneath the retinal pigment epithelium (RPE). Previous studies of SFD patient-induced pluripotent stem cell (iPSC) derived RPE differ as to whether these cultures recapitulate this key clinical feature by forming increased drusenoid deposits. The primary purpose of this study is to examine whether SFD patient-derived iPSC-RPE form basal deposits similar to what is found in affected family member SFD globes and to determine whether SFD iPSC RPE may be more oxidatively stressed. We performed a careful comparison of iPSC RPE from three control individuals, multiple iPSC clones from two SFD patients' iPSC RPE, and post-mortem eyes of affected SFD family members. We also examined the effect of CRISPR-Cas9 gene correction of the S204C TIMP3 mutation on RPE phenotype. Finally, targeted metabolomics with liquid chromatography and mass spectrometry analysis and stable isotope-labeled metabolite analysis were performed to determine whether SFD RPE are more oxidatively stressed. We found that SFD iPSC-RPE formed significantly more sub-RPE deposits (∼6-90 µm in height) compared to control RPE at 8 weeks. These deposits were similar in composition to the thick layer of sub-RPE deposits found in SFD family member globes by immunofluorescence staining and TEM imaging. S204C TIMP3 correction by CRISPR-Cas9 gene editing in SFD iPSC RPE cells resulted in significantly reduced basal laminar and sub-RPE calcium deposits. We detected a ∼18-fold increase in TIMP3 accumulation in the extracellular matrix (ECM) of SFD RPE, and targeted metabolomics showed that intracellular 4-hydroxyproline, a major breakdown product of collagen, is significantly elevated in SFD RPE, suggesting increased ECM turnover. Finally, SFD RPE cells have decreased intracellular reduced glutathione and were found to be more vulnerable to oxidative stress. Our findings suggest that elements of SFD pathology can be demonstrated in culture which may lead to insights into disease mechanisms.


Subject(s)
Induced Pluripotent Stem Cells , Macular Degeneration , Extracellular Matrix/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Macular Degeneration/metabolism , Retinal Pigment Epithelium/metabolism
3.
Int J Mol Sci ; 23(22)2022 Nov 17.
Article in English | MEDLINE | ID: mdl-36430707

ABSTRACT

Sorsby fundus dystrophy (SFD) is an autosomal dominant macular disorder caused by mutations in tissue Inhibitor of the metalloproteinase-3 (TIMP3) gene with the onset of symptoms including choroidal neovascularization as early as the second decade of life. We have previously reported that wild-type TIMP3 is an endogenous angiogenesis inhibitor that inhibits Vascular Endothelial Growth Factor (VEGF)-mediated signaling in endothelial cells. In contrast, SFD-related S179C-TIMP3 when expressed in endothelial cells, does not have angiogenesis-inhibitory properties. To evaluate if this is a common feature of TIMP3 mutants associated with SFD, we examined and compared endothelial cells expressing S179C, Y191C and S204C TIMP3 mutants for their angiogenesis-inhibitory function. Western blot analysis, zymography and reverse zymography and migration assays were utilized to evaluate TIMP3 protein, Matrix Metalloproteinase (MMP) and MMP inhibitory activity, VEGF signaling and in vitro migration in endothelial cells expressing (VEGF receptor-2 (VEGFR-2) and wild-type TIMP3 or mutant-TIMP3. We demonstrate that mutant S179C, Y191C- and S204C-TIMP3 all show increased glycosylation and multimerization/aggregation of the TIMP3 protein. In addition, endothelial cells expressing TIMP3 mutations show increased angiogenic activities and elevated VEGFR-2. Removal of N-glycosylation by mutation of Asn184, the only potential N-glycosylation site in mutant TIMP3, resulted in increased aggregation of TIMP3, further upregulation of VEGFR-2, VEGF-induced phosphorylation of VEGFR2 and VEGF-mediated migration concomitant with reduced MMP inhibitory activity. These results suggest that even though mutant TIMP3 proteins are more glycosylated, post-translational deglycosylation may play a critical role in the aggregation of mutant TIMP3 and contribute to the pathogenesis of SFD. The identification of factors that might contribute to changes in the glycome of patients with SFD will be useful. Future studies will evaluate whether variations in the glycosylation of mutant TIMP3 proteins are contributing to the severity of the disease.


Subject(s)
Macular Degeneration , Tissue Inhibitor of Metalloproteinase-3 , Vascular Endothelial Growth Factor Receptor-2 , Humans , Endothelial Cells/metabolism , Matrix Metalloproteinases/metabolism , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factors/metabolism , Glycosylation , Macular Degeneration/genetics , Macular Degeneration/metabolism , Protein Aggregates/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism
4.
Nanomedicine ; 28: 102205, 2020 08.
Article in English | MEDLINE | ID: mdl-32305594

ABSTRACT

Choroidal neovascularization (CNV) is the abnormal growth of blood vessels that sprout from the choroid vasculature and grow beneath and into the retina. The newly formed blood vessels in CNV often leak blood and fluid which deteriorates vision over time, eventually leading to blindness. In the present study, we examined the efficacy of intravenously injected gold nanoparticles in the laser-induced CNV animal model. Using optical coherence tomography (OCT) and fluorescein angiography, we evaluated CNV lesions longitudinally, over a period of 21 days, with and without nanoparticle treatment. Intravenously injected low concentration of bare gold nanoparticles showed significant anti-angiogenic properties by suppressing CNV development and progression. The treatment group showed significantly decreased fluorescein leakage at the CNV site compared to vehicle injected control mice. OCT assisted CNV volume measurement at all time points showed a significant reduction in lesion size in the treatment group compared with controls.


Subject(s)
Choroidal Neovascularization/drug therapy , Gold/chemistry , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Tomography, Optical Coherence/methods , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/therapeutic use , Animals , Female , Fluorescein Angiography/methods , Male , Mice
5.
J Neurosci Res ; 97(1): 88-97, 2019 01.
Article in English | MEDLINE | ID: mdl-30129971

ABSTRACT

Sorsby fundus dystrophy (SFD), an autosomal dominant, fully penetrant, degenerative disease of the macula, is manifested by symptoms of night blindness or sudden loss of visual acuity, usually in the third to fourth decades of life due to choroidal neovascularization (CNV). SFD is caused by specific mutations in the Tissue Inhibitor of Metalloproteinase-3, (TIMP3) gene. The predominant histo-pathological feature in the eyes of patients with SFD are confluent 20-30 m thick, amorphous deposits found between the basement membrane of the retinal pigment epithelium (RPE) and the inner collagenous layer of Bruch's membrane. SFD is a rare disease but it has generated significant interest because it closely resembles the exudative or "wet" form of the more common age-related macular degeneration (AMD). In addition, in both SFD and AMD donor eyes, sub-retinal deposits have been shown to accumulate TIMP3 protein. Understanding the molecular functions of wild-type and mutant TIMP3 will provide significant insights into the patho-physiology of SFD and perhaps AMD. This review summarizes the current knowledge on TIMP3 and how mutations in TIMP3 cause SFD to provide insights into how we can study this disease going forward. Findings from these studies could have potential therapeutic implications for both SFD and AMD.


Subject(s)
Macular Degeneration/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics , Animals , Humans , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mutation , Retina/metabolism , Retina/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Tissue Inhibitor of Metalloproteinase-3/metabolism
6.
Exp Eye Res ; 185: 107672, 2019 08.
Article in English | MEDLINE | ID: mdl-31128100

ABSTRACT

Retinal lesions in the posterior pole of laboratory mice occur due to native, developmental abnormalities or as a consequence of environmental or experimental conditions. In this study, we investigated the rate and extent of retinal lesions as a result of prolonged ocular exposure following general anesthesia. Following experimental preparation induction procedures (EPIP) involving general anesthesia, mydriasis/cycloplegia, and topical anesthesia to the cornea, two ocular recovery conditions (protected and unprotected) were tested within two different animal recovery chambers (open or closed). The anterior and posterior poles were evaluated for the development of retinal lesions using digital color photography, scanning laser ophthalmoscopy, and spectral-domain optical coherence during anesthesia recovery and up to 2.5 months thereafter. In some mice, electroretinograms, histological and immunohistological evaluations were performed to assess functional and structural changes that accompanied the retinal lesions detected by in vivo imaging. Our data suggests that prolonged ocular surface exposure to circulating ambient room air leads to significant anterior and posterior segment ocular complications. The most abundant, semi-reversible complication observed was the development of lesions in the outer retina, which had a 90% probability of occurring after 45 min of exposure. The lesions mostly resolved short-term, but functional and imaging evidence suggest that some perturbations to the outer retina may persist one or more months following initial development.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/adverse effects , Anesthetics, Combined/adverse effects , Anesthetics, Dissociative/adverse effects , Hypnotics and Sedatives/adverse effects , Retina/drug effects , Retinal Diseases/chemically induced , Animals , Biomarkers/metabolism , Color Vision/physiology , Electroretinography , Female , Fluorescein Angiography , Immunohistochemistry , Ketamine/adverse effects , Male , Mice , Mice, Inbred C57BL , Mydriatics/adverse effects , Night Vision/physiology , Ophthalmoscopy , Pentobarbital/adverse effects , Retina/metabolism , Retina/physiopathology , Retinal Diseases/metabolism , Retinal Diseases/physiopathology , Tomography, Optical Coherence , Xylazine/adverse effects
7.
Exp Eye Res ; 184: 30-37, 2019 07.
Article in English | MEDLINE | ID: mdl-30978346

ABSTRACT

A patient with bilateral diffuse uveal melanocytic proliferation (BDUMP) associated with endometrial cancer was treated with plasmapheresis, but failed therapy with progressive serous retinal detachment. We collected plasma before and after plasmapheresis therapy. Our goal was to determine if the cultured melanocyte elongation and proliferation (CMEP) factor and hepatocyte growth factor (HGF) was present in the IgG enriched fraction and understand why our patient failed plasmapheresis therapy. Melanocytes were cultured for 3-5 days in the presence of control medium, unfractionated pre-plasmapheresis BDUMP medium, IgG enriched or IgG depleted BDUMP medium, or unfractionated post-plasmapheresis BDUMP medium. Subretinal fluid was collected from patients with BDUMP and control retinal detachments and analyzed by electropheresis with immunoblotting. Medium with unfractionated BDUMP plasma stimulated melanocyte growth 1.4-1.5 fold compared to control medium on days 3-5 (p < 0.001 for all). Both IgG enriched and IgG depleted BDUMP medium mildly increased melanocyte growth 1.3 fold (p < 0.05 for enriched, p < 0.01 for depleted) compared to control. In comparison, unfractionated BDUMP medium caused a 1.7-fold increase in melanocyte growth, which was significantly more than the enriched (p < 0.01) and depleted (p < 0.05) fractions. Pre-plasmapheresis and post-plasmapheresis unfractionated BDUMP medium equally stimulated melanocyte growth 1.7-fold (p < 0.05) compared to control. HGF was present in IgG depleted, pre-plasmapheresis, and post-plasmapheresis samples, but absent in the IgG enriched fraction. There was no enrichment of IgG in the subretinal fluid from eyes with BDUMP. In conclusion, CMEP factor is not concentrated in the IgG enriched plasma fraction in our patient who failed plasmapheresis therapy. HGF levels have no correlation with melanocyte growth. Because plasmapheresis preferentially removes immunoglobulins from the plasma, our patient responded poorly to plasmapheresis treatment with worsening retinal detachment.


Subject(s)
Adenocarcinoma, Clear Cell/pathology , Endometrial Neoplasms/pathology , Intercellular Signaling Peptides and Proteins/blood , Melanocytes/pathology , Paraneoplastic Syndromes, Ocular/pathology , Uvea/pathology , Adenocarcinoma, Clear Cell/blood , Adenocarcinoma, Clear Cell/therapy , Aged , Cell Proliferation , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Endometrial Neoplasms/blood , Endometrial Neoplasms/therapy , Female , Fluorescein Angiography , Humans , Immunoblotting , Multimodal Imaging , Paraneoplastic Syndromes, Ocular/blood , Paraneoplastic Syndromes, Ocular/therapy , Plasmapheresis , Subretinal Fluid , Treatment Failure
8.
Exp Eye Res ; 180: 63-74, 2019 03.
Article in English | MEDLINE | ID: mdl-30543793

ABSTRACT

In diabetes, there are two major physiological aberrations: (i) Loss of insulin signaling due to absence of insulin (type 1 diabetes) or insulin resistance (type 2 diabetes) and (ii) increased blood glucose levels. The retina has a high proclivity to damage following diabetes, and much of the pathology seen in diabetic retinopathy has been ascribed to hyperglycemia and downstream cascades activated by increased blood glucose. However, less attention has been focused on the direct role of insulin on retinal physiology, likely due to the fact that uptake of glucose in retinal cells is not insulin-dependent. The retinal pigment epithelium (RPE) is instrumental in maintaining the structural and functional integrity of the retina. Recent studies have suggested that RPE dysfunction is a precursor of, and contributes to, the development of diabetic retinopathy. To evaluate the role of insulin on RPE cell function directly, we generated a RPE specific insulin receptor (IR) knockout (RPEIRKO) mouse using the Cre-loxP system. Using this mouse, we sought to determine the impact of insulin-mediated signaling in the RPE on retinal function under physiological control conditions as well as in streptozotocin (STZ)-induced diabetes. We demonstrate that loss of RPE-specific IR expression resulted in lower a- and b-wave electroretinogram amplitudes in diabetic mice as compared to diabetic mice that expressed IR on the RPE. Interestingly, RPEIRKO mice did not exhibit significant differences in the amplitude of the RPE-dependent electroretinogram c-wave as compared to diabetic controls. However, loss of IR-mediated signaling in the RPE reduced levels of reactive oxygen species and the expression of pro-inflammatory cytokines in the retina of diabetic mice. These results imply that IR-mediated signaling in the RPE regulates photoreceptor function and may play a role in the generation of oxidative stress and inflammation in the retina in diabetes.


Subject(s)
Diabetic Retinopathy/metabolism , Insulin/physiology , Retinal Pigment Epithelium/metabolism , Retinal Rod Photoreceptor Cells/physiology , Signal Transduction/physiology , Animals , Blood Glucose/metabolism , Blotting, Western , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Retinopathy/physiopathology , Electroretinography , Genetic Markers , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Retina/physiopathology
9.
FASEB J ; 32(10): 5674-5684, 2018 10.
Article in English | MEDLINE | ID: mdl-29874129

ABSTRACT

The predominant function of the blood-retinal barrier (BRB) is to maintain retinal homeostasis by regulating the influx and efflux between the blood and retina. Breakdown of the BRB occurs in a number of ocular diseases that result in vision loss. Understanding the molecular and cellular pathways involved in the development and maintenance of the BRB is critical to developing therapeutics for these conditions. To visualize the BRB in vivo, we used the transgenic Tg(l-fabp:DBP-EGFP:flk1:mCherry) zebrafish model that expresses vitamin D binding protein (a member of the albumin gene family) tagged to green fluorescent protein. Retinoic acid (RA) plays a number of important roles in vertebrate development and has been shown to play a protective role during inflammation-induced blood-brain barrier disruption. The role of RA in BRB development and maintenance remains unknown. To disrupt RA signaling, Tg(l-fabp:DBP-EGFP:flk1:mCherry) zebrafish were treated with N, N-diethylaminobenzaldehyde and 4-[(1 E)-2-[5,6-dihydro-5,5-dimethyl-8-(2-phenylethynyl)-2-naphthalenyl]ethenyl]benzoic acid, which are antagonists of retinal dehydrogenase and the RA receptor, respectively. Treatment with either compound resulted in BRB disruption and reduced visual acuity, whereas cotreatment with all- trans RA effectively rescued BRB integrity. Additionally, transgenic overexpression of Cyp26a1, which catalyzes RA degradation, resulted in breakdown of the BRB. Our results demonstrate that RA signaling is critical for maintenance of the BRB and could play a role in diseases such as diabetic macular edema.-Pollock, L. M., Xie, J., Bell, B. A., Anand-Apte, B. Retinoic acid signaling is essential for maintenance of the blood-retinal barrier.


Subject(s)
Blood-Retinal Barrier/metabolism , Signal Transduction/drug effects , Tretinoin , Zebrafish/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Blood-Retinal Barrier/pathology , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Retinoic Acid 4-Hydroxylase/genetics , Retinoic Acid 4-Hydroxylase/metabolism , Signal Transduction/genetics , Tretinoin/pharmacokinetics , Tretinoin/pharmacology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
10.
FASEB J ; 31(12): 5208-5216, 2017 12.
Article in English | MEDLINE | ID: mdl-28784632

ABSTRACT

Opioids modulate the tumor microenvironment with potential functional consequences for tumor growth and metastasis. We evaluated the effects of morphine administration on the circulating proteolytic profile of tumor-free mice. Serum from morphine-treated (1 or 10 mg/kg, i.p. every 12 h) or saline-treated mice was collected at different time points and tested ex vivo in endothelial, lymphatic endothelial, and breast cancer cell migration assays. Serum from mice that were treated with 10 mg/kg morphine for 3 d displayed reduced chemotactic potential for endothelial and breast cancer cells, and elicited reduced cancer cell invasion through reconstituted basement membrane compared with serum from saline controls. This was associated with decreased circulating matrix metalloproteinase 9 (MMP-9) and increased circulating tissue inhibitor of metalloproteinase 1 (TIMP-1) and TIMP-3/4 as assessed by zymography and reverse zymography. By using quantitative RT-PCR, we confirmed morphine-induced alterations in MMP-9 and TIMP expression and identified organs, including the liver and spleen, in which these changes originated. Pharmacologic inhibition of MMP-9 abrogated the difference in chemotactic attraction between serum from saline-treated and morphine-treated mice, which indicated that reduced proteolytic ability mediated the decreased migration toward serum from morphine-treated mice. This novel mechanism may enable morphine administration to promote an environment that is less conducive to tumor growth, invasion, and metastasis.-Xie, N., Khabbazi, S., Nassar, Z. D., Gregory, K., Vithanage, T., Anand-Apte, B., Cabot, P. J., Sturgess, D., Shaw, P. N., Parat, M.-O. Morphine alters the circulating proteolytic profile in mice: functional consequences on cellular migration and invasion.


Subject(s)
Cell Movement/drug effects , Matrix Metalloproteinase 9/metabolism , Morphine/pharmacology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , Analgesics, Opioid/pharmacology , Animals , Cattle , Cell Line , Cell Line, Tumor , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred BALB C , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics
11.
J Immunol ; 196(5): 2377-87, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26810221

ABSTRACT

Angiogenesis is closely linked to and precedes eosinophilic infiltration in asthma. Eosinophils are recruited into the airway by chemoattractant eotaxins, which are expressed by endothelial cells, smooth muscles cells, epithelial cells, and hematopoietic cells. We hypothesized that bone marrow-derived proangiogenic progenitor cells that contain eotaxins contribute to the initiation of angiogenesis and inflammation in asthma. Whole-lung allergen challenge of atopic asthma patients revealed vascular activation occurs within hours of challenge and before airway inflammation. The eotaxin receptor CCR3 was expressed at high levels on submucosal endothelial cells in patients and a murine model of asthma. Ex vivo exposure of murine endothelial cells to eotaxins induced migration and angiogenesis. In mechanistic studies, wild-type mice transplanted with eotaxin-1/2-deficient bone marrow had markedly less angiogenesis and inflammation in an atopic asthma model, whereas adoptive transfer of proangiogenic progenitor cells from wild-type mice in an atopic asthma model into the eotaxin-1/2-deficient mice led to angiogenesis and airway inflammation. The findings indicate that Th2-promoting hematopoietic progenitor cells are rapidly recruited to the lung upon allergen exposure and release eotaxins that coordinately activate endothelial cells, angiogenesis, and airway inflammation.


Subject(s)
Asthma/metabolism , Asthma/pathology , Chemokine CCL11/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Hematopoietic Stem Cells/metabolism , Neovascularization, Pathologic/metabolism , Receptors, CCR3/metabolism , Adoptive Transfer , Adult , Allergens/immunology , Animals , Asthma/genetics , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Case-Control Studies , Chemokine CCL11/genetics , Chemokine CCL24/genetics , Chemokine CCL24/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Eosinophils/immunology , Eosinophils/metabolism , Female , Humans , Hypersensitivity, Immediate/genetics , Hypersensitivity, Immediate/metabolism , Hypersensitivity, Immediate/pathology , Immunohistochemistry , Male , Mice , Mice, Knockout , Th2 Cells/immunology , Th2 Cells/metabolism
12.
Mol Vis ; 23: 140-148, 2017.
Article in English | MEDLINE | ID: mdl-28356706

ABSTRACT

PURPOSE: Familial exudative vitreoretinopathy (FEVR) is caused by mutations in the genes encoding low-density lipoprotein receptor-related protein (LRP5) or its interacting partners, namely frizzled class receptor 4 (FZD4) and norrin cystine knot growth factor (NDP). Mouse models for Lrp5, Fzd4, and Ndp have proven to be important for understanding the retinal pathophysiology underlying FEVR and systemic abnormalities related to defective Wnt signaling. Here, we report a new mouse mutant, tvrm111B, which was identified by electroretinogram (ERG) screening of mice generated in the Jackson Laboratory Translational Vision Research Models (TVRM) mutagenesis program. METHODS: ERGs were used to examine outer retinal physiology. The retinal vasculature was examined by in vivo retinal imaging, as well as by histology and immunohistochemistry. The tvrm111B locus was identified by genetic mapping of mice generated in a cross to DBA/2J, and subsequent sequencing analysis. Gene expression was examined by real-time PCR of retinal RNA. Bone mineral density (BMD) was examined by peripheral dual-energy X-ray absorptiometry. RESULTS: The tvrm111B allele is inherited as an autosomal recessive trait. Genetic mapping of the decreased ERG b-wave phenotype of tvrm111B mice localized the mutation to a region on chromosome 19 that included Lrp5. Sequencing of Lrp5 identified the insertion of a cytosine (c.4724_4725insC), which is predicted to cause a frameshift that disrupts the last three of five conserved PPPSPxS motifs in the cytoplasmic domain of LRP5, culminating in a premature termination. In addition to a reduced ERG b-wave, Lrp5tvrm111B homozygotes have low BMD and abnormal features of the retinal vasculature that have been reported previously in Lrp5 mutant mice, including persistent hyaloid vessels, leakage on fluorescein angiography, and an absence of the deep retinal capillary bed. CONCLUSIONS: The phenotype of the Lrp5tvrm111B mutant includes abnormalities of the retinal vasculature and of BMD. This model may be a useful resource to further our understanding of the biological role of LRP5 and to evaluate experimental therapies for FEVR or other conditions associated with LRP5 dysfunction.


Subject(s)
Bone Density , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Mutagenesis/genetics , Mutation/genetics , Retinal Vessels/abnormalities , Retinal Vessels/physiopathology , Animals , Electroretinography , Gene Expression Regulation , Homozygote , Male , Mice, Inbred C57BL , Organ Size/genetics , Phenotype , Retinal Vessels/diagnostic imaging , Retinal Vessels/pathology , Wnt Signaling Pathway/genetics
13.
Apoptosis ; 20(4): 523-34, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25558000

ABSTRACT

Tissue inhibitor of metalloproteinases-3 (TIMP3) is a tumor suppressor and a potent inhibitor of angiogenesis. TIMP3 exerts its anti-angiogenic effect via a direct interaction with vascular endothelial growth factor (VEGF) receptor-2 (KDR) and inhibition of proliferation, migration and tube formation of endothelial cells (ECs). TIMP3 has also been shown to induce apoptosis in some cancer cells and vascular smooth muscle cells via MMP inhibition and caspase-dependent mechanisms. In this study, we examined the molecular mechanisms of TIMP3-mediated apoptosis in endothelial cells. We have previously demonstrated that mice developed smaller tumors with decreased vascularity when injected with breast carcinoma cells overexpressing TIMP3, than with control breast carcinoma cells. TIMP3 overexpression resulted in increased apoptosis in human breast carcinoma (MDA-MB435) in vivo but not in vitro. However, TIMP3 could induce apoptosis in ECs in vitro. The apoptotic activity of TIMP3 in ECs appears to be independent of MMP inhibitory activity. Furthermore, the equivalent expression of functional TIMP3 promoted apoptosis and caspase activation in ECs expressing KDR (PAE/KDR), but not in ECs expressing PDGF beta-receptor (PAE/ß-R). Surprisingly, the apoptotic activity of TIMP3 appears to be independent of caspases. TIMP3 inhibited matrix-induced focal adhesion kinase (FAK) tyrosine phosphorylation and association with paxillin and disrupted the incorporation of ß3 integrin, FAK and paxillin into focal adhesion contacts on the matrix, which were not affected by caspase inhibitors. Thus, TIMP3 may induce apoptosis in ECs by triggering a caspase-independent cell death pathway and targeting a FAK-dependent survival pathway.


Subject(s)
Apoptosis , Caspases/metabolism , Endothelial Cells/cytology , Tissue Inhibitor of Metalloproteinase-3/metabolism , Animals , Cell Line , Endothelial Cells/metabolism , Humans , Mice , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , Swine , Tissue Inhibitor of Metalloproteinase-3/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
14.
Am J Physiol Renal Physiol ; 307(12): F1427-34, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25298528

ABSTRACT

The zebrafish pronephros is a well-established model to study glomerular development, structure, and function. A few methods have been described to evaluate glomerular barrier function in zebrafish larvae so far. However, there is a need to assess glomerular filtration as well. In the present study, we extended the available methods by simultaneously measuring the intravascular clearances of Alexa fluor 647-conjugated 10-kDa dextran and FITC-conjugated 500-kDa dextran as indicators of glomerular filtration and barrier function, respectively. After intravascular injection of the dextrans, mean fluorescence intensities of both dextrans were measured in the cardinal vein of living zebrafish (4 days postfertilization) by confocal microscopy over time. We demonstrated that injected 10-kDa dextran was rapidly cleared from the circulation, became visible in the lumen of the pronephric tubule, quickly accumulated in tubular cells, and was detectably excreted at the cloaca. In contrast, 500-kDa dextran could not be visualized in the tubule at any time point. To check whether alterations in glomerular function can be quantified by our method, we injected morpholino oligonucleotides (MOs) against zebrafish nonmuscle myosin heavy chain IIA (zMyh9) or apolipoprotein L1 (zApol1). While glomerular filtration was reduced in zebrafish nonmuscle myosin heavy chain IIA MO-injected larvae, glomerular barrier function remained intact. In contrast, in zebrafish apolipoprotein L1 MO-injected larvae, glomerular barrier function was compromised as 500-kDa dextran disappeared from the circulation and became visible in tubular cells. In summary, we present a novel method that allows to simultaneously assess glomerular filtration and barrier function in live zebrafish.


Subject(s)
Glomerular Filtration Barrier/metabolism , Glomerular Filtration Rate , Zebrafish/metabolism , Animals , Apolipoproteins/genetics , Apolipoproteins/metabolism , Carbocyanines/metabolism , Dextrans/blood , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescent Dyes/metabolism , Larva/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Morpholinos/administration & dosage , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Renal Elimination , Time Factors , Zebrafish/blood , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
15.
Am J Pathol ; 183(3): 987-95, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23831329

ABSTRACT

Diabetes mellitus is a disease with considerable morbidity and mortality worldwide. Breakdown of the blood-retinal barrier and leakage from the retinal vasculature leads to diabetic macular edema, an important cause of vision loss in patients with diabetes. Although epidemiologic studies and randomized clinical trials suggest that glycemic control plays a major role in the development of vascular complications of diabetes, insulin therapies for control of glucose metabolism cannot prevent long-term retinal complications. The phenomenon of temporary paradoxical worsening of diabetic macular edema after insulin treatment has been observed in a number of studies. In prospective studies on non-insulin-dependent (type 2) diabetes mellitus patients, a change in treatment from oral drugs to insulin was often associated with a significant increased risk of retinopathy progression and visual impairment. Although insulin therapies are critical for regulation of the metabolic disease, their role in the retina is controversial. In this study with diabetic mice, insulin treatment resulted in increased vascular leakage apparently mediated by betacellulin and signaling via the epidermal growth factor (EGF) receptor. In addition, treatment with EGF receptor inhibitors reduced retinal vascular leakage in diabetic mice on insulin. These findings provide unique insight into the role of insulin signaling in mediating retinal effects in diabetes and open new avenues for therapeutics to treat the retinal complications of diabetes mellitus.


Subject(s)
Blood Vessels/pathology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Epidermal Growth Factor/metabolism , Protective Agents/metabolism , Signal Transduction , ADAM Proteins/metabolism , ADAM10 Protein , Amyloid Precursor Protein Secretases/metabolism , Animals , Betacellulin , Blood Vessels/drug effects , Capillary Permeability/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Humans , Insulin/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Macular Edema/metabolism , Macular Edema/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Protein Kinase Inhibitors/pharmacology , Retina , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Signal Transduction/drug effects , Tight Junctions/metabolism
16.
Exp Eye Res ; 129: 107-18, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25447564

ABSTRACT

Over the past 3 decades the zebrafish (Danio rerio) has become an important biomedical research species. As their use continues to grow additional techniques and tools will be required to keep pace with ongoing research using this species. In this paper we describe a novel method for in vivo imaging of the retinal vasculature in adult animals using a commercially available confocal scanning laser ophthalmoscope (SLO). With this instrumentation, we demonstrate the ability to distinguish diverse vascular phenotypes in different transgenic GFP lines. In addition this technology allows repeated visualization of the vasculature in individual zebrafish over time to document vascular leakage progression and recovery induced by intraocular delivery of proteins that induce vascular permeability. SLO of the retinal vasculature was found to be highly informative, providing images of high contrast and resolution that were capable of resolving individual vascular endothelial cells. Finally, the procedures required to acquire SLO images from zebrafish are non-invasive, simple to perform and can be achieved with low animal mortality, allowing repeated imaging of individual fish.


Subject(s)
Capillary Permeability , Diagnostic Imaging/methods , Ophthalmoscopy/methods , Retinal Vessels/cytology , Animals , Fundus Oculi , Zebrafish
17.
Am J Pathol ; 180(4): 1702-14, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22322296

ABSTRACT

Caveolin-1 is an essential structural protein of caveolae, specialized plasma membrane organelles highly abundant in endothelial cells, where they regulate multiple functions including angiogenesis. Caveolin-1 exerts a tonic inhibition of endothelial nitric oxide synthase (eNOS) activity. Accordingly, caveolin-1 gene-disrupted mice have enhanced eNOS activity as well as increased systemic nitric oxide (NO) levels. We hypothesized that excess eNOS activity, secondary to caveolin deficiency, would mediate the decreased angiogenesis observed in caveolin-1 gene-disrupted mice. We tested tumor angiogenesis in mice lacking either one or both proteins, using in vitro, ex vivo, and in vivo assays. We show that endothelial cell migration, tube formation, cell sprouting from aortic rings, tumor growth, and angiogenesis are all significantly impaired in both caveolin-1-null and eNOS-null mice. We further show that these parameters were either partially or fully restored in double knockout mice that lack both caveolin-1 and eNOS. Furthermore, the effects of genetic ablation of eNOS are mimicked by the administration of the NOS inhibitor N-nitro-L-arginine methyl ester hydrochloride (L-NAME), including the reversal of the caveolin-1-null mouse angiogenic phenotype. This study is the first to demonstrate the detrimental effects of unregulated eNOS activity on angiogenesis, and shows that impaired tumor angiogenesis in caveolin-1-null mice is, at least in part, the result of enhanced eNOS activity.


Subject(s)
Caveolin 1/deficiency , Neovascularization, Pathologic/metabolism , Nitric Oxide Synthase Type III/physiology , Animals , Caveolin 1/genetics , Caveolin 1/physiology , Cells, Cultured , Chemotaxis/physiology , Corneal Neovascularization/chemically induced , Corneal Neovascularization/metabolism , Corneal Neovascularization/prevention & control , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/physiology , Enzyme Inhibitors/pharmacology , Lung/cytology , Lung/drug effects , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , NG-Nitroarginine Methyl Ester/therapeutic use , Neoplasm Transplantation , Neovascularization, Pathologic/prevention & control , Neovascularization, Physiologic/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Vascular Endothelial Growth Factor A/pharmacology
18.
Transl Vis Sci Technol ; 12(7): 6, 2023 07 03.
Article in English | MEDLINE | ID: mdl-37410472

ABSTRACT

Purpose: To develop and train a deep learning-based algorithm for detecting disorganization of retinal inner layers (DRIL) on optical coherence tomography (OCT) to screen a cohort of patients with diabetic retinopathy (DR). Methods: In this cross-sectional study, subjects over age 18, with ICD-9/10 diagnoses of type 2 diabetes with and without retinopathy and Cirrus HD-OCT imaging performed between January 2009 to September 2019 were included in this study. After inclusion and exclusion criteria were applied, a final total of 664 patients (5992 B-scans from 1201 eyes) were included for analysis. Five-line horizontal raster scans from Cirrus HD-OCT were obtained from the shared electronic health record. Two trained graders evaluated scans for presence of DRIL. A third physician grader arbitrated any disagreements. Of 5992 B-scans analyzed, 1397 scans (∼30%) demonstrated presence of DRIL. Graded scans were used to label training data for the convolution neural network (CNN) development and training. Results: On a single CPU system, the best performing CNN training took ∼35 mins. Labeled data were divided 90:10 for internal training/validation and external testing purpose. With this training, our deep learning network was able to predict the presence of DRIL in new OCT scans with a high accuracy of 88.3%, specificity of 90.0%, sensitivity of 82.9%, and Matthews correlation coefficient of 0.7. Conclusions: The present study demonstrates that a deep learning-based OCT classification algorithm can be used for rapid automated identification of DRIL. This developed tool can assist in screening for DRIL in both research and clinical decision-making settings. Translational Relevance: A deep learning algorithm can detect disorganization of retinal inner layers in OCT scans.


Subject(s)
Deep Learning , Diabetes Mellitus, Type 2 , Diabetic Retinopathy , Humans , Adolescent , Diabetic Retinopathy/diagnostic imaging , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/diagnostic imaging , Cross-Sectional Studies , Fluorescein Angiography/methods , Retrospective Studies , Visual Acuity , Biomarkers , Tomography, Optical Coherence/methods
19.
Ophthalmic Surg Lasers Imaging Retina ; 54(12): 692-700, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38113360

ABSTRACT

BACKGROUND AND OBJECTIVE: Disorganization of retinal inner layers (DRIL) is a potential spectral-domain optical coherence tomography (SD-OCT) imaging biomarker with clinical utility in diabetic retinopathy (DR). PATIENTS AND METHODS: A cross-sectional study was conducted at a large academic center. The cohort was composed of 1,175 patients with type 2 diabetes with and without retinopathy on initial examination between September 2009 and January 2019 (n = 2,083 eyes). DR risk and progression factors were obtained from the medical record. Trained graders masked to patients' clinical histories evaluated SD-OCT scans for DRIL. RESULTS: Of 2,083 eyes, 28.1% (n = 585) demonstrated presence of DRIL with high interrater reliability (K = 0.88, 95% CI 0.86-0.90). DRIL was associated with worse visual acuity (VA) (P < 0.001) and DR severity (P < 0.0001). Insulin users had more severe DR (P < 0.0001). DR-related factors, race (Black, White) and sex (male) were significantly associated with DRIL (P < 0.05). CONCLUSIONS: DRIL was strongly associated with DR severity and worse VA, supporting its utility as an unfavorable prognostic indicator. [Ophthalmic Surg Lasers Imaging Retina 2023;54:692-700.].


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Retinopathy , Macular Edema , Humans , Male , Diabetic Retinopathy/complications , Tomography, Optical Coherence/methods , Diabetes Mellitus, Type 2/complications , Cross-Sectional Studies , Reproducibility of Results , Macular Edema/diagnosis , Retrospective Studies , Fluorescein Angiography/methods , Retina
20.
J Biol Chem ; 286(26): 23511-20, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21543326

ABSTRACT

Angiogenesis requires concomitant remodeling of cell junctions and migration, as exemplified by recent observations of extensive endothelial cell movement along growing blood vessels. We report that a protein complex that regulates cell junctions is required for VEGF-driven directional migration and for angiogenesis in vivo. The complex consists of RhoA and Syx, a RhoA guanine exchange factor cross-linked by the Crumbs polarity protein Mupp1 to angiomotin, a phosphatidylinositol-binding protein. The Syx-associated complex translocates to the leading edge of migrating cells by membrane trafficking that requires the tight junction recycling GTPase Rab13. In turn, Rab13 associates with Grb2, targeting Syx and RhoA to Tyr(1175)-phosphorylated VEGFR2 at the leading edge. Rab13 knockdown in zebrafish impeded sprouting of intersegmental vessels and diminished the directionality of their tip cells. These results indicate that endothelial cell mobility in sprouting vessels is facilitated by shuttling the same protein complex from disassembling junctions to the leading edges of cells.


Subject(s)
Cell Movement/physiology , Endothelial Cells/metabolism , Neovascularization, Physiologic/physiology , rab GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Endothelial Cells/cytology , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Membrane Proteins , Mice , Mice, Knockout , Phosphorylation/physiology , Tight Junctions/genetics , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zebrafish/embryology , Zebrafish/genetics , rab GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL