Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
FASEB J ; 34(8): 10117-10131, 2020 08.
Article in English | MEDLINE | ID: mdl-32525594

ABSTRACT

Mast cells (MCs) are the initial responders of innate immunity and their degranulation contribute to various etiologies. While the abundance of MCs in the choroid implies their fundamental importance in the eye, little is known about the significance of MCs and their degranulation in choroid. The cause of geographic atrophy (GA), a progressive dry form of age-related macular degeneration is elusive and there is currently no therapy for this blinding disorder. Here we demonstrate in both human GA and a rat model for GA, that MC degranulation and MC-derived tryptase are central to disease progression. Retinal pigment epithelium degeneration followed by retinal and choroidal thinning, characteristic phenotypes of GA, were driven by continuous choroidal MC stimulation and activation in a slow release fashion in the rat. Genetic manipulation of MCs, pharmacological intervention targeting MC degranulation with ketotifen fumarate or inhibition of MC-derived tryptase with APC 366 prevented all of GA-like phenotypes following MC degranulation in the rat model. Our results demonstrate the fundamental role of choroidal MC involvement in GA disease etiology, and will provide new opportunities for understanding GA pathology and identifying novel therapies targeting MCs.


Subject(s)
Geographic Atrophy/pathology , Mast Cells/pathology , Animals , Cell Line , Choroid/metabolism , Choroid/pathology , Disease Models, Animal , Geographic Atrophy/metabolism , Humans , Macular Degeneration/metabolism , Macular Degeneration/pathology , Male , Mast Cells/metabolism , Rats , Rats, Sprague-Dawley , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Tryptases/metabolism
2.
Article in English | MEDLINE | ID: mdl-37992236

ABSTRACT

At the earliest stage of battery development, differential scanning calorimetry (DSC) of a sample with all battery cell stack materials can provide quantitative data on the reaction thermochemistry. The resulting quantitative thermochemical map of expected reactions upon heating can then guide chemistry and component development toward improved cell safety. In this work, we construct Li0.43CoO2 + C + PVDF|Li6.4La3Zr1.4Ta0.6O12|Li microcell DSC samples with capacity-matched electrodes and test to 500 °C. Notable observations are: (1) ∼74% of the O2 released from the Li0.43CoO2 cathode reacts with C to form CO2 rather than with molten Li to produce Li2O, (2) PVDF pyrolysis (>400 °C) releases HF gas that exothermically reacts with Li to form LiF, and (3) reactions involving oxygen (e.g., CO2 and Li2O formation) account for ∼60% of the total heat released, and reactions involving HF (e.g., LiF formation) account for ∼36% of the total heat released.

3.
Invest Ophthalmol Vis Sci ; 64(15): 39, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38153753

ABSTRACT

Purpose: Proliferative vitreoretinopathy (PVR) is the most common cause of failure of surgically repaired rhegmatogenous retinal detachment (RRD). Chemically induced and cell injection PVR models do not fully simulate the clinical characteristics of PVR in the post-RRD context. There is an unmet need for translational models in which to study mechanisms and treatments specific to RRD-PVR. Methods: RRD was induced in adult Dutch Belted rabbits. Posterior segments were fixed or processed for RNA sequencing at 6 hours and 2, 7, 14, and 35 days after induction. Histochemical staining and immunolabeling for glial fibrillary acidic protein, alpha smooth muscle actin, vascular endothelial growth factor receptor 2, CD68, and RPE 65 kDa protein were performed, and labeling intensity was scored. Single cell RNA sequencing was performed. Results: Acute histopathological changes included intravitreal and intraretinal hemorrhage, leukocytic vitritis, chorioretinitis, and retinal rarefaction. Chronic lesions showed retinal atrophy, gliosis, fibrotic subretinal membranes, and epiretinal fibrovascular proliferation. Fibrillar collagen was present in the fibrocellular and fibrovascular membranes in chronic lesions. Moderate to strong labeling of glia and vasculature was detected in chronic lesions. At day 14, most cells profiled by single cell sequencing were identified as Mϋller glia and microglia, consistent with immunolabeling. Expression of several fibrillar collagen genes was upregulated in chronic lesions. Conclusions: Histological and transcriptional features of this rabbit model simulate important features of human RRD-PVR, including the transition to chronic intraretinal and periretinal fibrosis. This animal model of RRD with features of PVR will enable further research on targeted treatment interventions.


Subject(s)
Retinal Detachment , Vitreoretinopathy, Proliferative , Adult , Animals , Humans , Rabbits , Vitreoretinopathy, Proliferative/etiology , Retinal Detachment/etiology , Vascular Endothelial Growth Factor A , Models, Animal , Fibrosis , Fibrillar Collagens
4.
Appl Opt ; 51(15): 2925-34, 2012 May 20.
Article in English | MEDLINE | ID: mdl-22614595

ABSTRACT

We present a study of Poincaré-beam polarization patterns produced by collinear superposition of two Laguerre-Gauss spatial modes in orthogonal polarization eigenstates (circular or linear). We explore theoretically and experimentally the combinations that are possible. We find that the resulting patterns can be explained in terms of mappings of points on the Poincaré sphere onto points in the transverse plane of the beam mode. The modes that we produced yielded many types of polarization singularities.

5.
Transl Vis Sci Technol ; 11(10): 36, 2022 10 03.
Article in English | MEDLINE | ID: mdl-36282118

ABSTRACT

Purpose: To evaluate the molecular, pharmacokinetic, and pharmacological properties of three anti-vascular endothelial growth factor (VEGF) agents-aflibercept, brolucizumab, and ranibizumab-and to provide a prediction of the optimal design of an intravitreal VEGF challenge in rabbits to assess the preclinical in vivo activity of the different anti-VEGF agents. Methods: Biochemical analyses and cellular and animal models of retinopathy were used to characterize anti-VEGF efficacy. Anti-VEGF biochemical binding affinity was determined through a kinetic exclusion assay. The in vitro potency was investigated by a calcium mobilization assay. Pharmacokinetic parameters were estimated for each drug to predict intraocular exposure relationships among the agents. The in silico modeling efforts informed the design of an in vivo rabbit model of VEGF-induced retinal hyperpermeability to determine the extent of VEGF neutralization in vivo. Consequently, data generated from the in vivo study enabled pharmacokinetic analysis and the generation of a logistical model describing the impact of the anti-VEGF agents on the VEGF-induced vascular leakage in rabbits. Results: The three anti-VEGF agents ranked from most efficacious to least efficacious as aflibercept, brolucizumab, and ranibizumab, with results consistent and significant within each individual characterization experiment. Conclusions: This composite study demonstrated how the molecular properties of aflibercept, brolucizumab, and ranibizumab translate into differences of in vivo efficacy, with results in line with the reported literature. Translational Relevance: In silico, in vitro, and in vivo integrated studies provide information that enables the enhanced characterization of translational properties of anti-VEGF agents currently used for the treatment of retinal diseases.


Subject(s)
Calcium , Ranibizumab , Animals , Rabbits , Ranibizumab/pharmacology , Ranibizumab/therapeutic use , Endothelial Growth Factors , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Intravitreal Injections , Vascular Endothelial Growth Factor A/metabolism
6.
J Cell Biol ; 161(5): 861-74, 2003 Jun 09.
Article in English | MEDLINE | ID: mdl-12796475

ABSTRACT

Nuclear factor of activated T cell (NFAT) is a ubiquitous regulator involved in multiple biological processes. Here, we demonstrate that NFAT is temporally required in the developing atrial myocardium between embryonic day 14 and P0 (birth). Inhibition of NFAT activity by conditional expression of dominant-negative NFAT causes thinning of the atrial myocardium. The thin myocardium exhibits severe sarcomere disorganization and reduced expression of cardiac troponin-I (cTnI) and cardiac troponin-T (cTnT). Promoter analysis indicates that NFAT binds to and regulates transcription of the cTnI and the cTnT genes. Thus, regulation of cytoskeletal protein gene expression by NFAT may be important for the structural architecture of the developing atrial myocardium.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Heart Atria/abnormalities , Heart Atria/growth & development , Myocardium/metabolism , Nuclear Proteins , Transcription Factors/metabolism , Active Transport, Cell Nucleus/genetics , Animals , Animals, Newborn , Binding Sites/genetics , Cell Nucleus/genetics , DNA-Binding Proteins/genetics , Down-Regulation/genetics , Fetus , Genes, Regulator/genetics , Heart Atria/metabolism , Mice , Mice, Transgenic , Microscopy, Electron , Mutation/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/ultrastructure , NFATC Transcription Factors , Promoter Regions, Genetic/genetics , Sarcomeres/metabolism , Sarcomeres/pathology , Sarcomeres/ultrastructure , Transcription Factors/genetics , Troponin I/biosynthesis , Troponin I/genetics , Troponin T/biosynthesis , Troponin T/genetics
7.
Invest Ophthalmol Vis Sci ; 48(3): 1212-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17325165

ABSTRACT

PURPOSE: Retinal ganglion cell (RGC) loss occurs in response to increased intraocular pressure (IOP) and/or retinal ischemia in glaucoma and leads to impairment of vision. This study was undertaken to test the efficacy of erythropoietin (EPO) in providing neuroprotection to RGCs in vivo. METHODS: The neuroprotective effects of EPO were studied in the DBA/2J mouse model of glaucoma. Mice were intraperitoneally injected with control substances or various doses of EPO, starting at the age of 6 months and continuing for an additional 2, 4, or 6 months. RGCs were labeled retrogradely by a gold tracer. IOP was measured with a microelectric-mechanical system, and EPO receptor (EPOR) expression was detected by immunohistochemistry. Axonal death in the optic nerve was quantified by para-phenylenediamine staining, and a complete blood count system was used to measure the number of erythrocytes. RESULTS: In DBA/2J mice, the average number of viable RGCs significantly decreased from 4 months to 10 months, with an inverse correlation between the number of dead optic nerve axons and viable RGCs. Treatment with EPO at doses of 3000, 6000, and 12,000 U/kg body weight per week all prevented significant RGC loss, compared with untreated DBA/2J control animals. EPO effects were similar to those of memantine, a known neuroprotective agent. IOP, in contrast, was unchanged by both EPO and memantine. Finally, EPOR was expressed in the RGC layer in both DBA/2J and C57BL/6J mice. CONCLUSIONS: EPO promoted RGC survival in DBA/2J glaucomatous mice without affecting IOP. These results suggest that EPO may be a potential therapeutic neuroprotectant in glaucoma.


Subject(s)
Cell Survival/drug effects , Erythropoietin/pharmacology , Glaucoma/prevention & control , Neuroprotective Agents/pharmacology , Optic Nerve Diseases/prevention & control , Retinal Ganglion Cells/drug effects , Animals , Axons/drug effects , Disease Models, Animal , Fluorescent Antibody Technique, Indirect , Glaucoma/metabolism , Glaucoma/pathology , Intraocular Pressure/drug effects , Memantine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Microscopy, Fluorescence , Optic Nerve/drug effects , Optic Nerve Diseases/metabolism , Optic Nerve Diseases/pathology , Receptors, Erythropoietin/metabolism , Recombinant Proteins , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
8.
Mol Cell Biol ; 22(11): 3905-26, 2002 Jun.
Article in English | MEDLINE | ID: mdl-11997523

ABSTRACT

The relationship between glycosylphosphatidyl inositol (GPI)-linked proteins and caveolins remains controversial. Here, we derived fibroblasts from Cav-1 null mouse embryos to study the behavior of GPI-linked proteins in the absence of caveolins. These cells lack morphological caveolae, do not express caveolin-1, and show a approximately 95% down-regulation in caveolin-2 expression; these cells also do not express caveolin-3, a muscle-specific caveolin family member. As such, these caveolin-deficient cells represent an ideal tool to study the role of caveolins in GPI-linked protein sorting. We show that in Cav-1 null cells GPI-linked proteins are preferentially retained in an intracellular compartment that we identify as the Golgi complex. This intracellular pool of GPI-linked proteins is not degraded and remains associated with intracellular lipid rafts as judged by its Triton insolubility. In contrast, GPI-linked proteins are transported to the plasma membrane in wild-type cells, as expected. Furthermore, recombinant expression of caveolin-1 or caveolin-3, but not caveolin-2, in Cav-1 null cells complements this phenotype and restores the cell surface expression of GPI-linked proteins. This is perhaps surprising, as GPI-linked proteins are confined to the exoplasmic leaflet of the membrane, while caveolins are cytoplasmically oriented membrane proteins. As caveolin-1 normally undergoes palmitoylation on three cysteine residues (133, 143, and 156), we speculated that palmitoylation might mechanistically couple caveolin-1 to GPI-linked proteins. In support of this hypothesis, we show that palmitoylation of caveolin-1 on residues 143 and 156, but not residue 133, is required to restore cell surface expression of GPI-linked proteins in this complementation assay. We also show that another lipid raft-associated protein, c-Src, is retained intracellularly in Cav-1 null cells. Thus, Golgi-associated caveolins and caveola-like vesicles could represent part of the transport machinery that is necessary for efficiently moving lipid rafts and their associated proteins from the trans-Golgi to the plasma membrane. In further support of these findings, GPI-linked proteins were also retained intracellularly in tissue samples derived from Cav-1 null mice (i.e., lung endothelial and renal epithelial cells) and Cav-3 null mice (skeletal muscle fibers).


Subject(s)
Caveolins/deficiency , Glycoproteins/metabolism , Glycosylphosphatidylinositols/metabolism , 3T3 Cells , Animals , Binding Sites , Biological Transport, Active , CSK Tyrosine-Protein Kinase , Caveolin 1 , Caveolins/chemistry , Caveolins/genetics , Caveolins/metabolism , Cell Compartmentation , Intracellular Fluid/metabolism , Kidney Tubules/metabolism , Lung/metabolism , Membrane Microdomains/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Palmitic Acid/metabolism , Protein-Tyrosine Kinases/metabolism , Transfection , src-Family Kinases
9.
Diabetes ; 54(3): 679-86, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15734843

ABSTRACT

Recently, we have shown that loss of caveolin-1 leads to marked alterations in insulin signaling and lipolysis in white adipose tissue. However, little is known about the role of caveolin-1 in brown adipose tissue (BAT), a tissue responsible for nonshivering thermogenesis. Here, we show that caveolin-1 null mice have a mildly, yet significantly, decreased resting core body temperature. To investigate this in detail, we next subjected the mice to fasting (for 24 h) or cold treatment (4 degrees C for 24 h), individually or in combination. Interestingly, caveolin-1 null mice showed markedly decreased body temperatures in response to fasting or fasting/cold treatment; however, cold treatment alone had no effect. In addition, under these conditions caveolin-1 null mice failed to show the normal increase in serum nonesterified fatty acids induced by fasting or fasting/cold treatment, suggesting that these mice are unable to liberate triglyceride stores for heat production. In accordance with these results, the triglyceride content of BAT was reduced nearly 10-fold in wild-type mice after fasting/cold treatment, but it was reduced only 3-fold in caveolin-1 null mice. Finally, electron microscopy of adipose tissue revealed dramatic perturbations in the mitochondria of caveolin-1 null interscapular brown adipocytes. Taken together, our data provide the first molecular genetic evidence that caveolin-1 plays a critical functional and structural role in the modulation of thermogenesis via an effect on lipid mobilization.


Subject(s)
Adipose Tissue, Brown/metabolism , Caveolins/physiology , Thermogenesis/physiology , Adipose Tissue, Brown/ultrastructure , Animals , Caveolin 1 , Caveolins/genetics , Cold Temperature , Fatty Acids, Nonesterified/blood , Food Deprivation/physiology , Gene Expression , Lipolysis/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/ultrastructure , Triglycerides/metabolism
10.
Cancer Biol Ther ; 5(3): 292-7, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16418573

ABSTRACT

Caveolin-1 (Cav-1) is the main structural protein of caveolae, plasma membrane invaginations that have been implicated in vesicular transport, cholesterol homeostasis, and the regulation of signal transduction. Previous in vivo studies have established a correlation between Cav-1 expression levels and milk production. In the normal mouse mammary gland, Cav-1 levels were shown to be downregulated during late pregnancy and lactation, via a Ras-p42/44-MAPK- dependent mechanism. Conversely, mammary glands from Cav-1 null-/- mice exhibit premature lactation, with augmented development of the lobulo-alveolar compartment and hyper-activation of the Jak-2/STAT5a signaling cascade. However, it remains unknown whether these phenotypes are cell-autonomous, i.e., intrinsic to the alveolar mammary epithelial cells, or whether stromal or adipocyte-secreted factors contribute. To directly address this issue, we have isolated primary mammary epithelial cells from wild-type (WT) and Cav-1 null-/- mammary glands. We cultured them either in a 2D model (monolayers of mammary epithelial cells) or in a 3D system on exogenous basement membrane (Matrigel; to reconstitute the minimal lactating unit, i.e., the mammary acinus). We show here that Cav-1 deficient mammary epithelial cells display the ability to spontaneously generate milk droplets, and to secrete them into the acinar lumen. Interestingly, such milk production occurs in the absence of lactogenic stimulation. Our results show that monolayers of Cav-1 null mammary epithelial cells are enriched in milk droplets, as judged by both (1) phase contrast microscopy and (2) immunofluorescence analysis with an antiserum directed against mouse milk proteins. Consistently, Cav-1 deficient mammary acini display increased milk production and secretion, as evaluated by Western blot analysis and electron microscopic examination. Mechanistically, we show that loss of Cav-1 in mammary epithelial cells induces the baseline constitutive hyper-activation of STAT5a signaling, which normally controls the temporal progression of lactogenesis in the mammary gland. The possible implications of our findings for understanding mammary tumorigenesis are also discussed.


Subject(s)
Caveolin 1/genetics , Epithelial Cells/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Milk/metabolism , STAT5 Transcription Factor/metabolism , Animals , Blotting, Western , Caveolin 1/physiology , Cell Culture Techniques , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Epithelial Cells/cytology , Female , Fluorescent Antibody Technique , Mice , Mice, Knockout , Signal Transduction
11.
Diabetes ; 53(5): 1261-70, 2004 May.
Article in English | MEDLINE | ID: mdl-15111495

ABSTRACT

Recently, it was shown that caveolin-1 can be redirected from the cell surface to intracellular lipid droplets in a variety of cell types. Here, we directly address the role of caveolin-1 in lipid droplet formation and breakdown, showing that caveolin-1 null mice exhibit markedly attenuated lipolytic activity. Mechanistically, although the activity of protein kinase A (PKA) was greatly increased in caveolin-1 null adipocytes, the phosphorylation of perilipin was dramatically reduced, indicating that caveolin-1 may facilitate the PKA-mediated phosphorylation of perilipin. In support of this hypothesis, coimmunoprecipitation experiments revealed that treatment with a beta(3)-adrenergic receptor agonist resulted in ligand-induced complex formation between perilipin, caveolin-1, and the catalytic subunit of PKA in wild-type but not in caveolin-1 null fat pads. We also show that caveolin-1 expression is important for efficient lipid droplet formation because caveolin-1 null embryonic fibroblasts stably transfected with perilipin accumulated approximately 4.5-fold less lipid than perilipin-transfected wild-type cells. Finally, high-pressure freeze-substitution electron microscopy of adipose tissue revealed dramatic perturbations in the architecture of the "lipid droplet cortex" (the interface between the lipid droplet surface and the cytoplasm) in caveolin-1 null perigonadal adipocytes. Taken together, our data provide the first molecular genetic evidence that caveolin-1 plays a critical functional and structural role in the modulation of both lipid droplet biogenesis and metabolism in vivo.


Subject(s)
Caveolins/physiology , Lipids/physiology , Lipolysis/physiology , Adipocytes/metabolism , Adipocytes/ultrastructure , Adipose Tissue/metabolism , Adrenergic beta-3 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Animals , Carrier Proteins , Caveolin 1 , Caveolins/deficiency , Caveolins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dioxoles/pharmacology , Fasting/metabolism , Fatty Acids, Nonesterified/blood , Lipid Metabolism , Mice , Mice, Knockout , Microscopy, Electron/methods , Perilipin-1 , Phosphoproteins/metabolism , Phosphorylation , Time Factors , Up-Regulation
12.
J Ocul Pharmacol Ther ; 29(8): 760-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23844757

ABSTRACT

PURPOSE: To evaluate the toxicokinetics and tolerability (local ocular and general toxicity) of the anti-inflammatory agent, dexamethasone phosphate (a prodrug of dexamethasone) delivered to the eye in rabbits by transscleral iontophoresis. METHODS: Female rabbits (n=6/group) received dexamethasone phosphate (40 mg/mL ophthalmic solution, EGP-437) transsclerally to the right eye (OD) using the Eyegate(®) II ocular iontophoresis delivery system once biweekly for 24 consecutive weeks at current doses of 10, 14, and 20 mA-min and current levels up to, and including -4 mA for 3.5-5 min. The study included 2 control groups (n=6/group): (1) a noniontophoresis control [an ocular applicator-loaded citrate buffer (placebo) without current] and (2) an iontophoresis control (a citrate buffer plus cathode iontophoresis at 20 mA-min, -4 mA for 5 min). Recoverability was evaluated 4 weeks following the last dose in 2 animals per group. The left eye (OS) was untreated and served as an internal control for each animal. Ocular and general safety of dexamethasone phosphate and dexamethasone were assessed. Other evaluations included toxicokinetics, ophthalmic examinations, intraocular pressure (IOP) measurements, electroretinographs, clinical observations, body weight, hematology and serum chemistry, gross necropsy, organ weight, and microscopic histopathology. RESULTS: The biweekly transscleral iontophoresis with either the citrate buffer or dexamethasone phosphate at cathodic doses up to, and including 20 mA-min and currents up to, and including -4 mA for 24 weeks was well-tolerated. Transient signs of conjunctival hyperemia and chemosis, mild corneal opacity, and fluorescein staining of the cornea were noted and attributed to expected ocular reactions to the temporary placement of the ocular applicator and application of iontophoresis. There were no dexamethasone phosphate-, dexamethasone-, or iontophoresis-related effects on IOP, electroretinography, or histopathology. Reductions in body weight gain, anemia, decreased leukocyte and lymphocyte counts, compromised liver function, enlarged liver, and reduced spleen weight were consistent with systemic corticosteroid-mediated pharmacology, repeated use of anesthesia, stress, and sedentariness, and unlikely to be related to iontophoresis application. CONCLUSIONS: The results of this investigation suggest that repeated transscleral iontophoresis with dexamethasone phosphate may be safe for use as a treatment for inflammatory ocular disorders that require prolonged and/or repeated corticosteroid therapy.


Subject(s)
Dexamethasone/analogs & derivatives , Drug Delivery Systems , Eye/drug effects , Glucocorticoids/adverse effects , Iontophoresis , Animals , Body Weight/drug effects , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Dexamethasone/blood , Dose-Response Relationship, Drug , Drug Administration Schedule , Eye/metabolism , Eye/pathology , Female , Glucocorticoids/administration & dosage , Glucocorticoids/blood , Ophthalmic Solutions , Organ Size/drug effects , Organ Specificity , Rabbits
13.
J Control Release ; 147(2): 225-31, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20655965

ABSTRACT

The fundamental understanding of ocular drug delivery using iontophoresis is not at the same level as that for transdermal electrotransport. Research has therefore been undertaken to characterise the electrical properties of the sclera (charge, permselectivity, and isoelectric point (pI)) and to determine the basics of iontophoretic transport of model neutral, cationic, and anionic species (respectively, mannitol, timolol, and dexamethasone phosphate). Like the skin, the sclera supports a net negative charge under physiological pH conditions and has a pI between 3.5 and 4. Equally, the principles of trans-scleral iontophoretic transport of low molecular weight compounds are consistent with those observed for skin. Iontophoretic delivery of timolol and dexamethasone phosphate was proportional to applied current and drug concentration, and trans-scleral iontophoresis in rabbits led to enhanced intraocular levels of these compounds compared to passive delivery. The behaviour of higher molecular weight species such as peptide drugs and other biopharmaceuticals (e.g., proteins and oligonucleotides) has not been fully characterised. Further work has been undertaken, therefore, to examine the trans-scleral iontophoresis of vancomycin, a glycopeptide antibiotic with a relatively high molecular weight of 1448 Da. It was indeed possible to deliver vancomycin by iontophoresis but trans-scleral transport did not increase linearly with either increasing current density or peptide concentration.


Subject(s)
Drug Delivery Systems/methods , Iontophoresis , Pharmaceutical Preparations/administration & dosage , Sclera/metabolism , Animals , Biological Transport , Chromatography, High Pressure Liquid , Dexamethasone/administration & dosage , Dexamethasone/analogs & derivatives , Dexamethasone/chemistry , Dexamethasone/pharmacokinetics , Drug Delivery Systems/instrumentation , Electrodes , Equipment Design , Female , In Vitro Techniques , Mannitol/administration & dosage , Mannitol/chemistry , Mannitol/pharmacokinetics , Molecular Weight , Pharmaceutical Preparations/chemistry , Rabbits , Tandem Mass Spectrometry , Timolol/administration & dosage , Timolol/chemistry , Timolol/pharmacokinetics , Vancomycin/administration & dosage , Vancomycin/chemistry , Vancomycin/pharmacokinetics
14.
J Biol Chem ; 282(38): 28045-56, 2007 Sep 21.
Article in English | MEDLINE | ID: mdl-17626017

ABSTRACT

The longer splice isoforms of vascular endothelial growth factor-A (VEGF-A), including mouse VEGF164, contain a highly basic heparin-binding domain (HBD), which imparts the ability of these isoforms to be deposited in the heparan sulfate-rich extracellular matrix and to interact with the prototype sulfated glycosaminoglycan, heparin. The shortest isoform, VEGF120, lacks this highly basic domain and is freely diffusible upon secretion. Although the HBD has been attributed significant relevance to VEGF-A biology, the molecular determinants of the heparin-binding site are unknown. We used site-directed mutagenesis to identify amino acid residues that are critical for heparin binding activity of the VEGF164 HBD. We focused on basic residues and found Arg-13, Arg-14, and Arg-49 to be critical for heparin binding and interaction with extracellular matrix in tissue samples. We also examined the cellular and biochemical consequences of abolishing heparin-binding function, measuring the ability of the mutants to interact with VEGF receptors, induce endothelial cell gene expression, and trigger microvessel outgrowth. Induction of tissue factor expression, vessel outgrowth, and binding to VEGFR2 were unaffected by the HBD mutations. In contrast, the HBD mutants showed slightly decreased binding to the NRP1 (neuropilin-1) receptor, and analyses suggested the heparin and NRP1 binding sites to be distinct but overlapping. Finally, mutations that affect the heparin binding activity also led to an unexpected reduction in the affinity of VEGF164 binding specifically to VEGFR1. This finding provides a potential basis for previous observations suggesting enhanced potency of VEGF164 versus VEGF120 in VEGFR1-mediated signaling in inflammatory cells.


Subject(s)
Heparin/chemistry , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/physiology , Amino Acid Sequence , Animals , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Mice , Molecular Sequence Data , Mutation , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Swine , Vascular Endothelial Growth Factor A/chemistry
15.
Dev Biol ; 303(2): 825-37, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17198697

ABSTRACT

Thin spongy myocardium is critical at early embryonic stage [before embryonic day (E) 13.5 in mice] to allow diffusion of oxygen and nutrients to the developing cardiomyocytes. However, establishment of compact myocardium at later stage ( approximately E16.5) during development is necessary to prepare for the increase in demand for blood circulation. Elucidating molecular targets of the spongy-compact myocardium transition between E13.5 and E16.5 in heart development is thus important. Previous studies demonstrated that multiple transcription factors and signaling pathways are involved in the regulation and function of the myocardium in heart development. Disruption of certain transcription factors or critical components of signaling pathways frequently causes structural malformation in heart and persistence of "thin spongy myocardium". We have recently demonstrated activation of the calcineurin/NFAT signaling pathway at E14.5 in developing myocardium. Constitutive inhibition of the calcineurin/NFAT signaling pathway caused embryonic lethality. Molecular targets downstream of the calcineurin/NFAT signaling pathway, however, remains elusive. Here, we report transcription targets, independently and dependently, regulated by the calcineurin/NFAT signaling during the E13.5-E16.5 myocardium transition. We have uncovered that expression of one-third of the induced genes during myocardium transition is calcineurin/NFAT-dependent. Among these calcineurin/NFAT-dependent transcription targets, there is a dosage-dependent regulation. Molecular studies indicate that formation of distinct NFAT:DNA complex, in part, accounts for the dosage-dependent regulation. Thus, in addition to temporal and spatial regulation, dosage-dependent threshold requirement provides another mechanism to modulate transcription response mediated by the calcineurin/NFAT signaling during heart development.


Subject(s)
Calcineurin/metabolism , Fetal Heart/embryology , Fetal Heart/metabolism , NFATC Transcription Factors/metabolism , Animals , Base Sequence , Calcineurin/genetics , DNA Primers/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , Pregnancy , Signal Transduction , Transcription, Genetic
16.
Am J Pathol ; 170(1): 316-33, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17200204

ABSTRACT

Here, we examine the role of "non-muscle" caveolins (Cav-1 and Cav-2) in skeletal muscle biology. Our results indicate that skeletal muscle fibers from male Cav-1(-/-) and Cav-2(-/-) mice show striking abnormalities, such as tubular aggregates, mitochondrial proliferation/aggregation, and increased numbers of M-cadherin-positive satellite cells. Notably, these skeletal muscle defects were more pronounced with increasing age. Because Cav-2-deficient mice displayed normal expression levels of Cav-1, whereas Cav-1-null mice exhibited an almost complete deficiency in Cav-2, these skeletal muscle abnormalities seem to be due to loss of Cav-2. Thus, Cav-2(-/-) mice represent a novel animal model-and the first genetically well-defined mouse model-that can be used to study the pathogenesis of tubular aggregate formation, which remains a poorly understood age-related skeletal muscle abnormality. Finally, because Cav-1 and Cav-2 were not expressed within mature skeletal myofibers, our results indicate that development of these abnormalities probably originates in stem/precursor cells, such as satellite cells or myoblasts. Consistent with this hypothesis, skeletal muscle isolated from male Cav-3(-/-) mice did not show any of these abnormalities. As such, this is the first study linking stem cells with the genesis of these intriguing muscle defects.


Subject(s)
Caveolin 1/genetics , Caveolin 2/genetics , Mitochondria, Muscle , Muscle Fibers, Skeletal , Muscular Diseases/genetics , Animals , Cadherins/biosynthesis , Caveolin 1/deficiency , Caveolin 2/deficiency , Disease Models, Animal , Electron Transport Complex IV/analysis , Genetic Predisposition to Disease , Male , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/ultrastructure , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/abnormalities , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Muscular Diseases/metabolism , Muscular Diseases/pathology , Myoblasts/metabolism , Myoblasts/pathology
17.
Am J Pathol ; 171(1): 53-67, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17591953

ABSTRACT

Vascular endothelial growth factor-A (VEGF-A) has recently been recognized as an important neuroprotectant in the central nervous system. Given its position as an anti-angiogenic target in the treatment of human diseases, understanding the extent of VEGF's role in neural cell survival is paramount. Here, we used a model of ischemia-reperfusion injury and found that VEGF-A exposure resulted in a dose-dependent reduction in retinal neuron apoptosis. Although mechanistic studies suggested that VEGF-A-induced volumetric blood flow to the retina may be partially responsible for the neuroprotection, ex vivo retinal culture demonstrated a direct neuroprotective effect for VEGF-A. VEGF receptor-2 (VEGFR2) expression was detected in several neuronal cell layers of the retina, and functional analyses showed that VEGFR2 was involved in retinal neuroprotection. VEGF-A was also shown to be involved in the adaptive response to retinal ischemia. Ischemic preconditioning 24 hours before ischemia-reperfusion injury increased VEGF-A levels and substantially decreased the number of apoptotic retinal cells. The protective effect of ischemic preconditioning was reversed after VEGF-A inhibition. Finally, chronic inhibition of VEGF-A function in normal adult animals led to a significant loss of retinal ganglion cells yet had no observable effect on several vascular parameters. These findings have implications for both neural pathologies and ocular vascular diseases, such as diabetic retinopathy and age-related macular degeneration.


Subject(s)
Reperfusion Injury/metabolism , Retina/physiology , Vascular Endothelial Growth Factor A/physiology , Adult , Animals , Apoptosis , Blood Flow Velocity , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Macular Degeneration , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Organ Culture Techniques , Rats , Rats, Long-Evans , Reperfusion Injury/pathology , Retina/drug effects , Retinal Vessels/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology
18.
Am J Pathol ; 168(1): 292-309, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16400031

ABSTRACT

During breast cancer development, the luminal space of the mammary acinar unit fills with proliferating epithelial cells that exhibit growth factor-independence, cell attachment defects, and a more invasive fibroblastic phenotype. Here, we used primary cultures of mammary epithelial cells derived from genetically engineered mice to identify caveolin-1 (Cav-1) as a critical factor for maintaining the normal architecture of the mammary acinar unit. Isolated cultures of normal mammary epithelial cells retained the capacity to generate mammary acini within extracellular matrix. However, those from Cav-1 (-/-) mice exhibited defects in three-dimensional acinar architecture, including disrupted lumen formation and epidermal growth factor-independent growth due to hyperactivation of the p42/44 mitogen-activated protein kinase cascade. In addition, Cav-1-null mammary epithelial cells deprived of exogenous extracellular matrix underwent a spontaneous epithelial-mesenchymal transition, with reorganization of the actin cytoskeleton, and E-cadherin redistribution. Mechanistically, these phenotypic changes appear to be caused by increases in matrix metalloproteinase-2/9 secretion and transforming growth factor-beta/Smad-2 hyperactivation. Finally, loss of Cav-1 potentiated the ability of growth factors (hepatocyte growth factor and basic fibroblast growth factor) to induce mammary acini branching, indicative of a more invasive fibroblastic phenotype. Thus, a Cav-1 deficiency profoundly affects mammary epithelia by modulating the activation state of important signaling cascades. Primary cultures of Cav-1-deficient mammary epithelia will provide a valuable new model to study the spatial/temporal progression of mammary cell transformation.


Subject(s)
Caveolin 1/deficiency , Cell Transformation, Neoplastic/ultrastructure , Mammary Glands, Animal/pathology , Precancerous Conditions/pathology , Signal Transduction/physiology , Animals , Blotting, Western , Cells, Cultured , Female , Fluorescent Antibody Technique , Growth Substances/metabolism , Mammary Glands, Animal/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Microscopy, Electron, Transmission , Neoplasm Invasiveness/pathology , Precancerous Conditions/metabolism , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism
19.
Am J Physiol Cell Physiol ; 288(3): C677-91, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15548572

ABSTRACT

It is generally well accepted that caveolin-3 expression is muscle specific, whereas caveolin-1 and -2 are coexpressed in a variety of cell types, including adipocytes, endothelial cells, epithelial cells, and fibroblasts. Caveolin-1 and -2 are known to form functional hetero-oligomeric complexes in cells where they are coexpressed, whereas caveolin-3 forms homo-oligomeric high molecular mass complexes. Although caveolin-2 might be expected to interact in a similar manner with caveolin-3, most studies indicate that this is not the case. However, this view has recently been challenged as it has been demonstrated that caveolin-2 and -3 are coexpressed in primary cultures of cardiac myocytes, where these two proteins can be coimmunoprecipitated. Thus it remains controversial whether caveolin-2 interacts with caveolin-3. Here, we directly address the issue of caveolin isoform protein-protein interactions by means of three distinct molecular genetic approaches. First, using caveolin-1-deficient mouse embryonic fibroblasts, in which we have stably expressed caveolin-1, -2, or -3, we find that caveolin-1 interacts with caveolin-2 in this setting, whereas caveolin-3 does not, in agreement with most published observations. Next, we used a transfected L6 myoblast cell system expressing all three caveolin proteins. Surprisingly, we found that caveolin-1, -2, and -3 all coimmunoprecipitate in this cell type, suggesting that this interaction is muscle cell specific. Similar results were obtained when the skeletal muscle of caveolin-1 transgenic animals was analyzed for caveolin-1 and caveolin-3 coimmunoprecipitation. Thus we conclude that all three caveolins can interact to form a discrete hetero-oligomeric complex, but that such complex formation is clearly muscle specific.


Subject(s)
Caveolins/metabolism , Fibroblasts/metabolism , Muscles/metabolism , Myoblasts/metabolism , Protein Isoforms/metabolism , Animals , Caveolae/chemistry , Caveolae/metabolism , Caveolins/genetics , Cell Line , Detergents/metabolism , Fibroblasts/ultrastructure , Macromolecular Substances , Mice , Mice, Knockout , Muscles/cytology , Myoblasts/ultrastructure , Octoxynol/metabolism , Protein Isoforms/genetics , Retroviridae/genetics , Retroviridae/metabolism
20.
Am J Physiol Cell Physiol ; 288(6): C1317-31, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15689413

ABSTRACT

Caveolin-3 (Cav-3) is expressed predominantly in skeletal muscle fibers, where it drives caveolae formation at the muscle cell's plasma membrane. In vitro studies have suggested that Cav-3 may play a positive role in insulin signaling and energy metabolism. We directly address the in vivo metabolic consequences of genetic ablation of Cav-3 in mice as it relates to insulin action, glucose metabolism, and lipid homeostasis. At age 2 mo, Cav-3 null mice are significantly larger than wild-type mice, and display significant postprandial hyperinsulinemia, whole body insulin resistance, and whole body glucose intolerance. Studies using hyperinsulinemic-euglycemic clamps revealed that Cav-3 null mice exhibited 20% and 40% decreases in insulin-stimulated whole body glucose uptake and whole body glycogen synthesis, respectively. Whole body insulin resistance was mostly attributed to 20% and 40% decreases in insulin-stimulated glucose uptake and glucose metabolic flux in the skeletal muscle of Cav-3 null mice. In addition, insulin-mediated suppression of hepatic glucose production was significantly reduced in Cav-3 null mice, indicating hepatic insulin resistance. Insulin-stimulated glucose uptake in white adipose tissue, which does not express Cav-3, was decreased by approximately 70% in Cav-3 null mice, suggestive of an insulin-resistant state for this tissue. During fasting, Cav-3 null mice possess normal insulin receptor protein levels in their skeletal muscle. However, after 15 min of acute insulin stimulation, Cav-3 null mice show dramatically reduced levels of the insulin receptor protein, compared with wild-type mice treated identically. These results suggest that Cav-3 normally functions to increase the stability of the insulin receptor at the plasma membrane, preventing its rapid degradation, i.e., by blocking or slowing ligand-induced receptor downregulation. Thus our results demonstrate the importance of Cav-3 in regulating whole body glucose homeostasis in vivo and its possible role in the development of insulin resistance. These findings may have clinical implications for the early diagnosis and treatment of caveolinopathies.


Subject(s)
Body Composition/physiology , Caveolins/physiology , Insulin Resistance/physiology , Muscle, Skeletal/physiology , Receptor, Insulin/physiology , Adipose Tissue/physiology , Animals , Blood Glucose/physiology , Caveolin 3 , Caveolins/genetics , Gene Expression , Glycogen/metabolism , Insulin/blood , Islets of Langerhans/pathology , Liver/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Receptor, Insulin/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL