Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Nat Commun ; 15(1): 8757, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39384746

ABSTRACT

Proliferative vitreoretinopathy is a vision-threatening response to penetrating ocular injury, for which there is no satisfactory treatment. In this disorder, retinal pigment epithelial cells, abandon their attachment to Bruch's membrane on the scleral side of the retina, transform into motile fibroblast-like cells, and migrate through the retinal wound to the vitreal surface of the retina, where they secrete membrane-forming proteins. Annexin A2 is a calcium-regulated protein that, in complex with S100A10, assembles plasmin-forming proteins at cell surfaces. Here, we show that, in proliferative vitreoretinopathy, recruitment of macrophages and directed migration of retinal pigment epithelial cells are annexin A2-dependent, and stimulated by macrophage inflammatory protein-1α/ß. These factors induce translocation of annexin A2 to the cell surface, thus enabling retinal pigment epithelial cell migration following injury; our studies reveal further that treatment of mice with intraocular antibody to either annexin A2 or macrophage inflammatory protein dampens the development of proliferative vitreoretinopathy in mice.


Subject(s)
Annexin A2 , Cell Movement , Macrophages , Retinal Pigment Epithelium , Vitreoretinopathy, Proliferative , Animals , Female , Humans , Male , Mice , Annexin A2/metabolism , Annexin A2/genetics , Inflammation/metabolism , Inflammation/pathology , Macrophages/metabolism , Macrophages/immunology , Mice, Inbred C57BL , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Vitreoretinopathy, Proliferative/metabolism , Vitreoretinopathy, Proliferative/pathology
2.
Invest Ophthalmol Vis Sci ; 64(4): 33, 2023 04 03.
Article in English | MEDLINE | ID: mdl-37103008

ABSTRACT

Purpose: We examined the role of annexin A2 (A2) in the development of diabetic retinal vasculopathy by testing the effect of Anxa2 gene deletion as well as administration of anti-A2 antibodies on pericyte dropout and retinal neovascularization in diabetic Akita mice, and in mice subjected to oxygen-induced retinopathy. Methods: We analyzed diabetic Ins2AKITA mice with or without global deletion of Anxa2, as well as Ins2AKITA mice that received intravitreal anti-A2 IgG or control antibody at 2, 4, and 6 months, for retinal pericyte dropout at 7 months of age. In addition, we assessed the effect of intravitreal anti-A2 on oxygen-induced retinopathy (OIR) in neonatal mice by quantifying retinal neovascular and vaso-obliterative area, and by enumeration of neovascular tufts. Results: Both deletion of the Anxa2 gene and immunologic blockade of A2 prevented pericyte depletion in retinas of diabetic Ins2AKITA mice. Blockade of A2 also reduced vaso-obliteration and neovascularization in the OIR model of vascular proliferation. This effect was amplified when a combination of antivascular endothelial growth factor (VEGF) and anti-A2 antibodies was used. Conclusions: Therapeutic approaches that target A2, alone or in combination with anti-VEGF therapy, are effective in mice, and may also curtail the progression of retinal vascular disease in humans with diabetes.


Subject(s)
Annexin A2 , Diabetes Mellitus , Diabetic Retinopathy , Retinal Diseases , Retinal Neovascularization , Animals , Mice , Annexin A2/genetics , Annexin A2/metabolism , Annexin A2/therapeutic use , Diabetic Retinopathy/prevention & control , Disease Models, Animal , Mice, Inbred C57BL , Oxygen/toxicity , Oxygen/metabolism , Retinal Diseases/metabolism , Retinal Neovascularization/metabolism , Vascular Endothelial Growth Factor A/metabolism
3.
Crit Care Explor ; 5(2): e0862, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36798534

ABSTRACT

The primary objective of this study was to determine whether expression of the multifunctional and adherens junction-regulating protein, annexin A2 (A2), is altered following cardiopulmonary bypass (CPB). A secondary objective was to determine whether depletion of A2 is associated with post-CPB organ dysfunction in children. DESIGN: In a prospective, observational study conducted over a 1-year period in children undergoing cardiac surgery requiring CPB, we analyzed A2 expression in peripheral blood mononuclear cells at different time points. We then assessed the relationship of A2 expression with organ function at each time point in the early postoperative period. SETTING: Twenty-three-bed mixed PICU in a tertiary academic center. PARTICIPANTS: Patients 1 month to 18 years old undergoing cardiac surgery requiring CPB. MEAN OUTCOME MEASUREMENTS AND RESULTS: We analyzed A2 expression in 22 enrolled subjects (n = 9, 1-23 mo old; n = 13, 2-18 yr old) and found a proteolysis-mediated decline in intact A2 immediately after bypass (p = 0.0009), reaching a median of 4% of baseline at 6 hours after bypass (p < 0.0001), and recovery by postoperative day 1. The degree of A2 depletion immediately after bypass in 1-23-month-olds correlated strongly with the extent of organ dysfunction, as measured by PICU admission Vasoactive-Ventilation-Renal (p = 0.004) and PEdiatric Logistic Organ Dysfunction-2 (p = 0.039) scores on postoperative day 1. A2 depletion immediately after bypass also correlated with more protracted requirement for both respiratory support (p = 0.007) and invasive ventilation (p = 0.013) in the 1-23-month-olds. CONCLUSIONS AND RELEVANCE: The degree of depletion of A2 following CPB correlates with more severe organ dysfunction, especially acute respiratory compromise in children under 2 years. These findings suggest that loss of A2 may contribute to pulmonary microvascular leak in young children following CPB.

4.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34782457

ABSTRACT

Lipofuscin granules enclose mixtures of cross-linked proteins and lipids in proportions that depend on the tissue analyzed. Retinal lipofuscin is unique in that it contains mostly lipids with very little proteins. However, retinal lipofuscin also presents biological and physicochemical characteristics indistinguishable from conventional granules, including indigestibility, tendency to cause lysosome swelling that results in rupture or defective functions, and ability to trigger NLRP3 inflammation, a symptom of low-level disruption of lysosomes. In addition, like conventional lipofuscins, it appears as an autofluorescent pigment, considered toxic waste, and a biomarker of aging. Ocular lipofuscin accumulates in the retinal pigment epithelium (RPE), whereby it interferes with the support of the neuroretina. RPE cell death is the primary cause of blindness in the most prevalent incurable genetic and age-related human disorders, Stargardt disease and age-related macular degeneration (AMD), respectively. Although retinal lipofuscin is directly linked to the cell death of the RPE in Stargardt, the extent to which it contributes to AMD is a matter of debate. Nonetheless, the number of AMD clinical trials that target lipofuscin formation speaks for the potential relevance for AMD as well. Here, we show that retinal lipofuscin triggers an atypical necroptotic cascade, amenable to pharmacological intervention. This pathway is distinct from canonic necroptosis and is instead dependent on the destabilization of lysosomes. We also provide evidence that necroptosis is activated in aged human retinas with AMD. Overall, this cytotoxicity mechanism may offer therapeutic targets and markers for genetic and age-related diseases associated with lipofuscin buildups.


Subject(s)
Intracellular Membranes/metabolism , Lipofuscin/pharmacology , Lysosomes/metabolism , Necroptosis/drug effects , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Aging , Alcohol Oxidoreductases , Animals , Cell Death , Humans , Lipofuscin/metabolism , Macular Degeneration/metabolism , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Retina/metabolism , Retinal Pigment Epithelium/metabolism
5.
J Exp Med ; 214(9): 2535-2545, 2017 Sep 04.
Article in English | MEDLINE | ID: mdl-28694388

ABSTRACT

Relative or absolute hypoxia activates signaling pathways that alter gene expression and stabilize the pulmonary microvasculature. Alveolar hypoxia occurs in disorders ranging from altitude sickness to airway obstruction, apnea, and atelectasis. Here, we report that the phospholipid-binding protein, annexin A2 (ANXA2) functions to maintain vascular integrity in the face of alveolar hypoxia. We demonstrate that microvascular endothelial cells (ECs) from Anxa2-/- mice display reduced barrier function and excessive Src-related tyrosine phosphorylation of the adherens junction protein vascular endothelial cadherin (VEC). Moreover, unlike Anxa2+/+ controls, Anxa2-/- mice develop pulmonary edema and neutrophil infiltration in the lung parenchyma in response to subacute alveolar hypoxia. Mice deficient in the ANXA2-binding partner, S100A10, failed to demonstrate hypoxia-induced pulmonary edema under the same conditions. Further analyses reveal that ANXA2 forms a complex with VEC and its phosphatases, EC-specific protein tyrosine phosphatase (VE-PTP) and Src homology phosphatase 2 (SHP2), both of which are implicated in vascular integrity. In the absence of ANXA2, VEC is hyperphosphorylated at tyrosine 731 in response to vascular endothelial growth factor, which likely contributes to hypoxia-induced extravasation of fluid and leukocytes. We conclude that ANXA2 contributes to pulmonary microvascular integrity by enabling VEC-related phosphatase activity, thereby preventing vascular leak during alveolar hypoxia.


Subject(s)
Annexin A2/physiology , Antigens, CD/physiology , Cadherins/physiology , Lung/blood supply , Microvessels/physiology , Protein Tyrosine Phosphatases/physiology , Animals , Annexin A2/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Female , Hypoxia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Edema/physiopathology , Vascular Endothelial Growth Factor A/physiology
6.
J Cell Sci ; 127(Pt 4): 828-44, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24357721

ABSTRACT

The annexins are an evolutionarily conserved family of phospholipid-binding proteins of largely unknown function. We observed that the AnxA2(-/-) lung basement membrane specifically lacks collagen VI (COL6), and postulated that ANXA2 directs bronchial epithelial cell secretion of COL6, an unusually large multimeric protein. COL6 serves to anchor cells to basement membranes and, unlike other collagens, undergoes multimerization prior to secretion. Here, we show that AnxA2(-/-) mice have reduced exercise tolerance with impaired lung tissue elasticity, which was phenocopied in Col6a1(-/-) mice. In vitro, AnxA2(-/-) fibroblasts retained COL6 within intracellular vesicles and adhered poorly to their matrix unless ANXA2 expression was restored. In vivo, AnxA2(-/-) bronchial epithelial cells underwent apoptosis and disadhesion. Immunoprecipitation and immunoelectron microscopy revealed that ANXA2 associates with COL6 and the SNARE proteins SNAP-23 and VAMP2 at secretory vesicle membranes of bronchial epithelial cells, and that absence of ANXA2 leads to retention of COL6 in a late-Golgi, VAMP2-positive compartment. These results define a new role for ANXA2 in the COL6 secretion pathway, and further show that this pathway establishes cell-matrix interactions that underlie normal pulmonary function and epithelial cell survival.


Subject(s)
Annexin A2/physiology , Apoptosis , Collagen Type VI/metabolism , Epithelial Cells/metabolism , Animals , Basement Membrane , Bronchi/metabolism , Bronchi/pathology , Cell Shape , Cell Survival , Cells, Cultured , Collagen Type VI/genetics , Elasticity , Epithelial Cells/physiology , Exercise Tolerance , Golgi Apparatus/metabolism , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Physical Exertion , Protein Transport , Pulmonary Ventilation , Respiratory Mucosa/pathology , Vesicle-Associated Membrane Protein 2/metabolism
7.
Nat Cell Biol ; 15(7): 807-17, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23728425

ABSTRACT

In a significant fraction of breast cancer patients, distant metastases emerge after years or even decades of latency. How disseminated tumour cells (DTCs) are kept dormant, and what wakes them up, are fundamental problems in tumour biology. To address these questions, we used metastasis assays in mice and showed that dormant DTCs reside on microvasculature of lung, bone marrow and brain. We then engineered organotypic microvascular niches to determine whether endothelial cells directly influence breast cancer cell (BCC) growth. These models demonstrated that endothelial-derived thrombospondin-1 induces sustained BCC quiescence. This suppressive cue was lost in sprouting neovasculature; time-lapse analysis showed that sprouting vessels not only permit, but accelerate BCC outgrowth. We confirmed this surprising result in dormancy models and in zebrafish, and identified active TGF-ß1 and periostin as tumour-promoting factors derived from endothelial tip cells. Our work reveals that stable microvasculature constitutes a dormant niche, whereas sprouting neovasculature sparks micrometastatic outgrowth.


Subject(s)
Bone Marrow Neoplasms/secondary , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Endothelium, Vascular/pathology , Lung Neoplasms/secondary , Neoplasm, Residual/pathology , Neovascularization, Pathologic , Pericytes/pathology , Animals , Bone Marrow Neoplasms/blood supply , Bone Marrow Neoplasms/metabolism , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Cell Adhesion Molecules/metabolism , Endothelium, Vascular/metabolism , Female , Fluorescent Antibody Technique , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/metabolism , Mice , Neoplasm, Residual/blood supply , Neoplasm, Residual/metabolism , Pericytes/metabolism , Stem Cell Niche/physiology , Thrombospondin 1/metabolism , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured , Tumor Microenvironment , Zebrafish/growth & development , Zebrafish/metabolism
8.
Blood ; 118(10): 2918-29, 2011 Sep 08.
Article in English | MEDLINE | ID: mdl-21788340

ABSTRACT

Oxygen-induced retinopathy (OIR) is a well-characterized model for retinopathy of prematurity, a disorder that results from rapid microvascular proliferation after exposure of the retina to high oxygen levels. Here, we report that the proliferative phase of OIR requires transcriptional induction of the annexin A2 (A2) gene through the direct action of the hypoxia-inducible factor-1 complex. We show, in addition, that A2 stabilizes its binding partner, p11, and promotes OIR-related angiogenesis by enabling clearance of perivascular fibrin. Adenoviral-mediated restoration of A2 expression restores neovascularization in the oxygen-primed Anxa2(-/-) retina and reinstates plasmin generation and directed migration in cultured Anxa2(-/-) endothelial cells. Systemic depletion of fibrin repairs the neovascular response to high oxygen treatment in the Anxa2(-/-) retina, whereas inhibition of plasminogen activation dampens angiogenesis under the same conditions. These findings show that the A2 system enables retinal neoangiogenesis in OIR by enhancing perivascular activation of plasmin and remodeling of fibrin. These data suggest new potential approaches to retinal angiogenic disorders on the basis of modulation of perivascular fibrinolysis.


Subject(s)
Annexin A2/physiology , Fibrin/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neovascularization, Pathologic , Oxygen/adverse effects , Retinal Diseases/etiology , Retinal Diseases/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Blotting, Western , Cell Movement , Cells, Cultured , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Fibrinolysin/metabolism , Fibrinolysis , Flow Cytometry , Gene Expression Regulation , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoenzyme Techniques , Immunoprecipitation , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasminogen Activators/metabolism , Promoter Regions, Genetic/genetics , RNA Stability , RNA, Messenger/genetics , Retinal Diseases/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Umbilical Veins/cytology , Umbilical Veins/metabolism
9.
J Clin Invest ; 119(11): 3384-94, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19841537

ABSTRACT

When plasma levels of homocysteine (HC), a thiol amino acid formed upon methionine demethylation, exceed 12 muM, individuals are at increased risk of developing large vessel atherothrombosis and small vessel dysfunction. The annexin A2 complex (termed "A2") is the cell surface coreceptor for plasminogen and TPA and accelerates the catalytic activation of plasmin, the major fibrinolytic agent in mammals. We previously showed that HC prevents A2-mediated, TPA-dependent activation of plasminogen in vitro by disulfide derivatization of the "tail" domain of A2. We also demonstrated that fibrinolysis and angiogenesis are severely impaired in A2-deficient mice. We now report here that, although hyperhomocysteinemic mice had a normal coagulation profile and normal platelet function, fibrin accumulated in their tissues due to reduced perivascular fibrinolytic activity and angiogenesis was impaired. A2 isolated from hyperhomocysteinemic mice failed to fully support TPA-dependent plasmin activation. However, infusion of hyperhomocysteinemic mice with fresh recombinant A2, which localized to neoangiogenic endothelial cells, resulted in normalization of angiogenesis and disappearance of peri- and intravascular fibrin. We therefore conclude that hyperhomocysteinemia impairs postnatal angiogenesis by derivatizing A2, preventing perivascular fibrinolysis, and inhibiting directed endothelial cell migration. These findings provide a mechanistic explanation for microvascular dysfunction and macrovascular occlusion in individuals with hyperhomocysteinemia.


Subject(s)
Annexin A2/metabolism , Fibrinolysis/physiology , Homocysteine/metabolism , Neovascularization, Physiologic/physiology , Animals , Annexin A2/pharmacology , Cell Movement/drug effects , Corneal Neovascularization/physiopathology , Diet , Endothelial Cells/cytology , Endothelial Cells/drug effects , Fibrin/metabolism , Fibrinolysis/drug effects , Homocysteine/pharmacology , Hyperhomocysteinemia/physiopathology , Male , Methionine/administration & dosage , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Recombinant Proteins/pharmacology
10.
Cell ; 137(6): 1018-31, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19524506

ABSTRACT

Retinoblastomas result from the inactivation of the RB1 gene and the loss of Rb protein, yet the cell type in which Rb suppresses retinoblastoma and the circuitry that underlies the need for Rb are undefined. Here, we show that retinoblastoma cells express markers of postmitotic cone precursors but not markers of other retinal cell types. We also demonstrate that human cone precursors prominently express MDM2 and N-Myc, that retinoblastoma cells require both of these proteins for proliferation and survival, and that MDM2 is needed to suppress ARF-induced apoptosis in cultured retinoblastoma cells. Interestingly, retinoblastoma cell MDM2 expression was regulated by the cone-specific RXRgamma transcription factor and a human-specific RXRgamma consensus binding site, and proliferation required RXRgamma, as well as the cone-specific thyroid hormone receptor-beta2. These findings provide support for a cone precursor origin of retinoblastoma and suggest that human cone-specific signaling circuitry sensitizes to the oncogenic effects of RB1 mutations.


Subject(s)
Cell Proliferation , Proto-Oncogene Proteins c-mdm2/metabolism , Retinoblastoma/metabolism , Signal Transduction , Animals , Cell Survival , Humans , Mice , Neoplasm Transplantation , Proto-Oncogene Proteins c-myc/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinoid X Receptor gamma/metabolism , Thyroid Hormone Receptors beta/metabolism , Transplantation, Heterologous
11.
Proc Natl Acad Sci U S A ; 104(29): 12005-10, 2007 Jul 17.
Article in English | MEDLINE | ID: mdl-17620600

ABSTRACT

The integrin receptor alphavbeta5 controls two independent forms of interactions of the retinal pigment epithelium (RPE) with adjacent photoreceptor outer segments that are essential for vision. Alphavbeta5 localizes specifically to apical microvilli of the RPE and contributes to retinal adhesion that maintains RPE contacts with intact outer segments at all times. Additionally, alphavbeta5 synchronizes diurnal bursts of RPE phagocytosis that clear photoreceptor outer segment fragments (POS) shed in a circadian rhythm. Dependence of retinal phagocytosis and adhesion on alphavbeta5 receptors suggests that the extracellular matrix ensheathing RPE microvilli contains ligands for this integrin. Here we studied mice lacking expression of functional MFG-E8 to test the contribution of this integrin ligand to alphavbeta5 functions in the retina. Lack of MFG-E8 only minimally reduced retinal adhesion. In contrast, lack of MFG-E8, like lack of alphavbeta5 receptor, eliminated alphavbeta5 downstream signaling involving the engulfment receptor MerTK and peak POS phagocytosis, both of which follow light onset in wild-type retina. MFG-E8-deficient RPE in primary culture retained normal epithelial morphology and levels of apical alphavbeta5 receptors, but showed impaired binding and engulfment of isolated POS. Soluble or POS-bound recombinant MFG-E8 was sufficient to fully restore phagocytosis by MFG-E8-deficient RPE. Furthermore, MFG-E8 supplementation strongly increased POS binding by wild-type and MerTK-deficient RPE, but did not affect POS binding by RPE lacking alphavbeta5. Thus, MFG-E8 stimulates rhythmic POS phagocytosis by ligating apical alphavbeta5 receptors of the RPE. These results identify MFG-E8 as the first extracellular ligand in the retina that is essential for diurnal POS phagocytosis.


Subject(s)
Antigens, Surface/metabolism , Circadian Rhythm , Integrins/metabolism , Milk Proteins/metabolism , Phagocytosis , Receptors, Vitronectin/metabolism , Rod Cell Outer Segment/cytology , Rod Cell Outer Segment/metabolism , Animals , Cell Adhesion , Enzyme Activation , Ligands , Mice , Mutant Proteins/metabolism , Phenotype , Pigment Epithelium of Eye/cytology , Protein Binding , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Recombinant Proteins/metabolism , Solubility , c-Mer Tyrosine Kinase
12.
Invest Ophthalmol Vis Sci ; 47(12): 5590-8, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17122153

ABSTRACT

PURPOSE: To define the pattern of Rb expression relative to cell cycle position and cell type in the developing human retina. METHODS: Cryosections of fetal week 11-18 retinas were immunostained for Rb and cell cycle- or cell type-specific markers. RESULTS: Rb was prominent in retinal progenitor cells (RPCs) expressing the cyclin D1, cyclin A, and cytoplasmic cyclin B markers of G1, S, and early to mid G2 phases, but not in RPCs expressing the phosphohistone H3 marker of late G2 and M. Rb was not detected in the earliest postmitotic ganglion, amacrine, horizontal, and bipolar cell precursors migrating away from the ventricular layer, but was detected as such cells underwent further differentiation. Among photoreceptors, Rb was not detected in the earliest RXRgamma(+) cone precursors or in the earliest Nrl(+) rod precursors, but subsequently rose to high levels in cones and to low levels in rods. Rb was prominent at the time when Müller glia exit the cell cycle and was generally expressed in a pattern complementary to p27(Kip1). CONCLUSIONS: Rb exhibits cell cycle-specific expression in RPCs, with loss in late G2-M and restoration in G1. Rb is re-expressed after postmitotic ganglion, amacrine, horizontal, and bipolar cell precursors migrate away from the ventricular layer; after the appearance of early cone and rod markers; but coinciding with Müller glia cell cycle withdrawal. The results suggest that Rb does not mediate the initial proliferative arrest of retinal neurons, but may indirectly induce arrest in RPCs or maintain an arrest in postmitotic precursors.


Subject(s)
Cell Cycle/physiology , Retina/embryology , Retinoblastoma Protein/metabolism , Amacrine Cells/cytology , Amacrine Cells/metabolism , Cyclin A/metabolism , Cyclin B/metabolism , Cyclin D , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclins/metabolism , Fluorescent Antibody Technique, Indirect , Gestational Age , Humans , Microscopy, Confocal , Phenotype , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/metabolism , Retina/cytology , Retina/metabolism , Retinal Bipolar Cells/cytology , Retinal Bipolar Cells/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Retinal Horizontal Cells/cytology , Retinal Horizontal Cells/metabolism , Stem Cells/cytology , Stem Cells/metabolism
13.
Arch Ophthalmol ; 121(1): 73-5, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12523888

ABSTRACT

OBJECTIVES: To quantify and analyze the differences between the length of the optic nerve as measured by the ophthalmologist in the operating room after enucleation and the length as measured by the pathologist after fixation. METHODS: The authors performed a retrospective review of patients who underwent either primary or secondary enucleation for retinoblastoma at the Ophthalmic Oncology Center of the New York-Presbyterian Hospital-Cornell campus between November 1979 and August 2001. Intraoperative notes and pathologic reports were reviewed to determine the length of the resected optic nerve as recorded by both the surgeon and pathologist. RESULTS: Sufficient data for inclusion in the study were available from 100 enucleation specimens belonging to 96 patients. A significant degree of shrinkage of the optic nerve occurred after fixation, with a mean shrinkage of 30.3% from the time of enucleation to the time of measurement by the pathologist. Age at enucleation affected the degree of optic nerve shrinkage; nerves from younger children underwent more shrinkage than nerves from older patients. Sex of the patient and the laterality of disease did not significantly affect optic nerve shrinkage. CONCLUSIONS: A significant degree of shrinkage of the optic nerve occurs in retinoblastoma enucleation specimens after fixation prior to pathologic analysis. This finding must be taken into account when comparing different series and making recommendations for chemoprophylaxis based solely on histopathologic examination.


Subject(s)
Eye Enucleation , Optic Nerve/pathology , Retinal Neoplasms/surgery , Retinoblastoma/surgery , Tissue Fixation , Child , Child, Preschool , Female , Humans , Infant , Male , Retrospective Studies , Tissue Embedding
SELECTION OF CITATIONS
SEARCH DETAIL