Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Pathol ; 193(11): 1789-1808, 2023 11.
Article in English | MEDLINE | ID: mdl-36965774

ABSTRACT

This study investigated retinal changes in a Western diet (WD)-induced nonhuman primate model of type 2 diabetes. Rhesus nonhuman primates, aged 15 to 17 years, were fed a high-fat diet (n = 7) for >5 years reflective of the traditional WD. Age-matched controls (n = 6) were fed a standard laboratory primate diet. Retinal fundus photography, optical coherence tomography, autofluorescence imaging, and fluorescein angiography were performed before euthanasia. To assess diabetic retinopathy (DR), eyes were examined using trypsin digests, lipofuscin autofluorescence, and multimarker immunofluorescence on cross-sections and whole mounts. Retinal imaging showed venous engorgement and tortuosity, aneurysms, macular exudates, dot and blot hemorrhages, and a marked increase in fundus autofluorescence. Post-mortem changes included the following: decreased CD31 blood vessel density (P < 0.05); increased acellular capillaries (P < 0.05); increased density of ionized calcium-binding adaptor molecule expressing amoeboid microglia/macrophage; loss of regular distribution in stratum and spacing typical of ramified microglia; and increased immunoreactivity of aquaporin 4 and glial fibrillary acidic protein (P < 0.05). However, rhodopsin immunoreactivity (P < 0.05) in rods and neuronal nuclei antibody-positive neuronal density of 50% (P < 0.05) were decreased. This is the first report of a primate model of DR solely induced by a WD that replicates key features of human DR.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Retinopathy , Animals , Humans , Diabetic Retinopathy/metabolism , Retinal Pigment Epithelium/metabolism , Diabetes Mellitus, Type 2/complications , Diet, Western , Retinal Vessels/metabolism , Primates , Tomography, Optical Coherence/methods
2.
Diabetologia ; 62(2): 322-334, 2019 02.
Article in English | MEDLINE | ID: mdl-30443753

ABSTRACT

AIMS/HYPOTHESIS: A major feature of diabetic retinopathy is breakdown of the blood-retinal barrier, resulting in macular oedema. We have developed a novel oligonucleotide-based drug, CD5-2, that specifically increases expression of the key junctional protein involved in barrier integrity in endothelial cells, vascular-endothelial-specific cadherin (VE-cadherin). CD5-2 prevents the mRNA silencing by the pro-angiogenic microRNA, miR-27a. CD5-2 was evaluated in animal models of ocular neovascularisation and vascular leak to determine its potential efficacy for diabetic retinopathy. METHODS: CD5-2 was tested in three mouse models of retinal dysfunction: conditional Müller cell depletion, streptozotocin-induced diabetes and oxygen-induced retinopathy. Vascular permeability in the Müller cell-knockout model was assessed by fluorescein angiography. The Evans Blue leakage method was used to determine vascular permeability in streptozotocin- and oxygen-induced retinopathy models. The effects of CD5-2 on retinal neovascularisation, inter-endothelial junctions and pericyte coverage in streptozotocin- and oxygen-induced retinopathy models were determined by staining for isolectin-B4, VE-cadherin and neural/glial antigen 2 (NG2). Blockmir CD5-2 localisation in diseased retina was determined using fluorescent in situ hybridisation. The effects of CD5-2 on VE-cadherin expression and in diabetic retinopathy-associated pathways, such as the transforming growth factor beta (TGF-ß) and wingless/integrated (WNT) pathway, were confirmed using western blot of lysates from HUVECs, a mouse brain endothelial cell line and a VE-cadherin null mouse endothelial cell line. RESULTS: CD5-2 penetrated the vasculature of the eye in the oxygen-induced retinopathy model. Treatment of diseased mice with CD5-2 resulted in reduced vascular leak in all three animal models, enhanced expression of VE-cadherin in the microvessels of the eye and improved pericyte coverage of the retinal vasculature in streptozotocin-induced diabetic models and oxygen-induced retinopathy models. Further, CD5-2 reduced the activation of retinal microglial cells in the streptozotocin-induced diabetic model. The positive effects of CD5-2 seen in vivo were further confirmed in vitro by increased protein expression of VE-cadherin, SMAD2/3 activity, and platelet-derived growth factor B (PDGF-B). CONCLUSIONS/INTERPRETATION: CD5-2 has therapeutic potential for individuals with vascular-leak-associated retinal diseases based on its ease of delivery and its ability to reverse vascular dysfunction and inflammatory aspects in three animal models of retinopathy.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetic Retinopathy/drug therapy , Oligonucleotides/therapeutic use , Animals , Blood-Retinal Barrier/metabolism , Capillary Permeability , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Mice , Retina/metabolism , Retinal Vessels/metabolism
3.
Stem Cells ; 36(11): 1764-1777, 2018 11.
Article in English | MEDLINE | ID: mdl-30068016

ABSTRACT

Identifying the signaling mechanisms that regulate adult neurogenesis is essential to understanding how the brain may respond to neuro-inflammatory events. P2X7 receptors can regulate pro-inflammatory responses, and in addition to their role as cation channels they can trigger cell death and mediate phagocytosis. How P2X7 receptors may regulate adult neurogenesis is currently unclear. Here, neural progenitor cells (NPCs) derived from adult murine hippocampal subgranular (SGZ) and cerebral subventricular (SVZ) zones were utilized to characterize the roles of P2X7 in adult neurogenesis, and assess the effects of high extracellular ATP, characteristic of inflammation, on NPCs. Immunocytochemistry found NPCs in vivo and in vitro expressed P2X7, and the activity of P2X7 in culture was demonstrated using calcium influx and pore formation assays. Live cell and confocal microscopy, in conjunction with flow cytometry, revealed P2X7+ NPCs were able to phagocytose fluorescent beads, and this was inhibited by ATP, indicative of P2X7 involvement. Furthermore, P2X7 receptors were activated with ATP or BzATP, and 5-ethynyl-2'-deoxyuridine (EdU) used to observe a dose-dependent decrease in NPC proliferation. A role for P2X7 in decreased NPC proliferation was confirmed using chemical inhibition and NPCs from P2X7-/- mice. Together, these data present three distinct roles for P2X7 during adult neurogenesis, depending on extracellular ATP concentrations: (a) P2X7 receptors can form transmembrane pores leading to cell death, (b) P2X7 receptors can regulate rates of proliferation, likely via calcium signaling, and (c) P2X7 can function as scavenger receptors in the absence of ATP, allowing NPCs to phagocytose apoptotic NPCs during neurogenesis. Stem Cells 2018;36:1764-1777.


Subject(s)
Hippocampus/metabolism , Inflammation/metabolism , Neural Stem Cells/metabolism , Receptors, Purinergic P2X7/metabolism , Stem Cells/metabolism , Animals , Cell Proliferation/physiology , Mice , Neural Stem Cells/cytology , Neurogenesis , Phagocytosis
4.
Stem Cells ; 33(2): 526-41, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25336287

ABSTRACT

During early human neurogenesis there is overproduction of neuroblasts and neurons accompanied by widespread programmed cell death (PCD). While it is understood that CD68(+) microglia and astrocytes mediate phagocytosis during target-dependent PCD, little is known of the cell identity or the scavenger molecules used to remove apoptotic corpses during the earliest stages of human neurogenesis. Using a combination of multiple-marker immunohistochemical staining, functional blocking antibodies and antagonists, we showed that human neural precursor cells (hNPCs) and neuroblasts express functional P2X7 receptors. Furthermore, using live-cell imaging, flow cytometry, phagocytic assays, and siRNA knockdown, we showed that in a serum-free environment, doublecortin(+) (DCX) neuroblasts and hNPCs can clear apoptotic cells by innate phagocytosis mediated via P2X7. We found that both P2X7(high) DCX(low) hNPCs and P2X7(high) DCX(high) neuroblasts, derived from primary cultures of human fetal telencephalon, phagocytosed targets including latex beads, apoptotic ReNcells, and apoptotic hNPC/neuroblasts. Pretreatment of neuroblasts and hNPCs with 1 mM adenosine triphosphate (ATP), 100 µM OxATP (P2X7 antagonist), or siRNA knockdown of P2X7 inhibited phagocytosis of these targets. Our results show that P2X7 functions as a scavenger receptor under serum-free conditions resembling those in early neurogenesis. This is the first demonstration that hNPCs and neuroblasts may participate in clearance of apoptotic corpses during pre target-dependent neurogenesis and mediate phagocytosis using P2X7 as a scavenger receptor.


Subject(s)
Fetus/metabolism , Neural Stem Cells/metabolism , Phagocytosis/physiology , Receptors, Purinergic P2X7/metabolism , Telencephalon/metabolism , Apoptosis/physiology , Cells, Cultured , Fetus/cytology , Gene Knockdown Techniques , Humans , Neural Stem Cells/cytology , Receptors, Purinergic P2X7/genetics , Telencephalon/cytology
5.
Am J Physiol Endocrinol Metab ; 308(8): E688-98, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25714673

ABSTRACT

Neuroinflammation and neurodegeneration have been observed in the brain in type 1 diabetes (T1D). However, little is known about the mediators of these effects. In T1D mice with 12- and 35-wk duration of diabetes we examined two mechanisms of neurodegeneration, loss of the neuroprotective factors insulin-like growth factor I (IGF-I) and IGF-binding protein-3 (IGFBP-3) and changes in indoleamine 2,3-dioxygenase (IDO) expression in the brain, and compared the response to age-matched controls. Furthermore, levels of matrix metalloproteinase-2 (MMP-2), nucleoside triphosphate diphosphohydrolase-1 (CD39), and ionized calcium-binding adaptor molecule 1 (Iba-1) were utilized to assess inflammatory changes in astrocytes, microglia, and blood vessels. In the diabetic hypothalamus (HYPO), we observed 20% reduction in neuronal soma diameter (P<0.05) and reduced neuronal expression of IGFBP-3 (-32%, P<0.05) and IGF-I (-15%, P<0.05) compared with controls at 35 wk. In diabetic HYPO, MMP-2 expression was increased in astrocytes (46%, P<0.01), and IDO⁺ cell density rose by (62%, P<0.05). CD39 expression dropped by 30% (P<0.05) in microglia and blood vessels. With 10 wk of systemic treatment using minocycline, an anti-inflammatory agent that crosses the blood-brain barrier, MMP-2, IDO, and CD39 levels normalized (P<0.05). Our results suggest that increased IDO and early loss of CD39⁺ protective cells lead to activation of inflammation in sympathetic centers of the CNS. As a downstream effect, the loss of the neuronal survival factors IGFBP-3 and IGF-I and the neurotoxic products of the kynurenine pathway contribute to the loss of neuronal density observed in the HYPO in T1D.


Subject(s)
Diabetes Mellitus, Type 1/complications , Diabetic Neuropathies/metabolism , Down-Regulation , Encephalitis/complications , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Sympathetic Nervous System/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/metabolism , Astrocytes/pathology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/physiopathology , Diabetic Neuropathies/immunology , Diabetic Neuropathies/pathology , Diabetic Neuropathies/prevention & control , Disease Progression , Down-Regulation/drug effects , Encephalitis/immunology , Encephalitis/metabolism , Encephalitis/prevention & control , Hypothalamus/drug effects , Hypothalamus/immunology , Hypothalamus/pathology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Minocycline/therapeutic use , Nerve Tissue Proteins/antagonists & inhibitors , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/immunology , Sympathetic Nervous System/pathology , Up-Regulation/drug effects
6.
Am J Pathol ; 183(5): 1608-20, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24160325

ABSTRACT

By using pseudorabies virus expressing green fluorescence protein, we found that efferent bone marrow-neural connections trace to sympathetic centers of the central nervous system in normal mice. However, this was markedly reduced in type 1 diabetes, suggesting a significant loss of bone marrow innervation. This loss of innervation was associated with a change in hematopoiesis toward generation of more monocytes and an altered diurnal release of monocytes in rodents and patients with type 1 diabetes. In the hypothalamus and granular insular cortex of mice with type 1 diabetes, bone marrow-derived microglia/macrophages were activated and found at a greater density than in controls. Infiltration of CD45(+)/CCR2(+)/GR-1(+)/Iba-1(+) bone marrow-derived monocytes into the hypothalamus could be mitigated by treatment with minocycline, an anti-inflammatory agent capable of crossing the blood-brain barrier. Our studies suggest that targeting central inflammation may facilitate management of microvascular complications.


Subject(s)
Bone Marrow/innervation , Bone Marrow/pathology , Central Nervous System/pathology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/pathology , Inflammation/pathology , Animals , Bone Marrow/drug effects , Central Nervous System/drug effects , Central Nervous System/metabolism , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Green Fluorescent Proteins/metabolism , Hematopoiesis/drug effects , Herpesvirus 1, Suid/drug effects , Herpesvirus 1, Suid/physiology , Humans , Inflammation/complications , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Minocycline/pharmacology , Models, Biological , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurotransmitter Agents/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/pathology
7.
Biogerontology ; 15(2): 177-98, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24337988

ABSTRACT

Over the last decade, the importance of NAD(+) has expanded beyond its role as an essential cofactor for energy metabolism. NAD(+) has emerged as a major signalling molecule that serves as the sole substrate for several enzymatic reactions including the DNA repair enzyme, poly(ADP-ribose) polymerase (PARP), NAD-dependent protein deacetylases or CD38, and transcriptional factors by a new class of histone deacetylases known as sirtuins. NAD(+) levels are regulated by the metabolic status and cellular stress caused by oxidative stress and DNA damage. Since a detailed study of NAD(+) metabolism in the healthy ageing mammalian brain is nascent, we examined the effect of ageing on intracellular NAD(+) metabolism in different brain regions in female Wistar rats in young (3 months), middle aged (12 months) and older adults (24 months). Our results are the first to show a significant decline in intracellular NAD(+) levels and NAD:NADH ratio with ageing in the CNS, occurring in parallel to an increase in lipid peroxidation and protein oxidation (o- and m-tyrosine) and a decline in total antioxidant capacity. Hyperphosphorylation of H2AX levels was also observed together with increased PARP-1 and PARP-2 expression, and CD38 activity, concomitantly with reduced NAD(+) and ATP levels and SIRT1 function in the cortex, brainstem, hippocampus and cerebellum. Reduced activity of mitochondrial complex I-IV and impaired maximum mitochondrial respiration rate were also observed in the ageing rat brain. Among the multiple physiological pathways associated with NAD(+) catabolism, our discovery of CD38 as the major regulator of cellular NAD(+) levels in rat neurons indicates that CD38 is a promising therapeutic target for the treatment of age-related neurodegenerative diseases.


Subject(s)
Aging/metabolism , Brain/metabolism , NAD/metabolism , ADP-ribosyl Cyclase/antagonists & inhibitors , ADP-ribosyl Cyclase/genetics , ADP-ribosyl Cyclase/metabolism , ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/metabolism , Adenosine Diphosphate Ribose/metabolism , Adenosine Triphosphate/biosynthesis , Animals , DNA Damage , Electron Transport , Female , Gene Knockdown Techniques , Lipid Peroxidation , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mitochondria/metabolism , Oxidative Stress , Poly(ADP-ribose) Polymerases/metabolism , Protein Carbonylation , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Sirtuin 1/metabolism , Tissue Distribution
8.
Invest Ophthalmol Vis Sci ; 65(4): 32, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38648041

ABSTRACT

Purpose: To undertake the first ultrastructural characterization of human retinal pigment epithelial (RPE) differentiation from fetal development to adolescence. Methods: Ten fetal eyes and three eyes aged six, nine, and 17 years were examined in the temporal retina adjacent to the optic nerve head by transmission electron microscopy. The area, number, and distribution of RPE organelles were quantified and interpreted within the context of adjacent photoreceptors, Bruch's membrane, and choriocapillaris maturation. Results: Between eight to 12 weeks' gestation (WG), pseudostratified columnar epithelia with apical tight junctions differentiate to a simple cuboidal epithelium with random distribution of melanosomes and mitochondria. Between 12 to 26 WG, cells enlarge and show long apical microvilli and apicolateral junctional complexes. Coinciding with eye opening at 26 WG, melanosomes migrate apically whereas mitochondria distribute to perinuclear regions, with the first appearance of phagosomes, complex granules, and basolateral extracellular space (BES) formation. Significantly, autophagy and heterophagy, as evidenced by organelle recycling, and the gold standard of ultrastructural evidence for autophagy of double-membrane autophagosomes and mitophagosomes were evident from 32 WG, followed by basal infoldings of RPE cell membrane at 36 WG. Lipofuscin formation and deposition into the BES evident at six years increased at 17 years. Conclusions: We provide compelling ultrastructural evidence that heterophagy and autophagy begins in the third trimester of human fetal development and that deposition of cellular byproducts into the extracellular space of RPE takes place via exocytosis. Transplanted RPE cells must also demonstrate the capacity to subserve autophagic and heterophagic functions for effective disease mitigation.


Subject(s)
Autophagy , Exocytosis , Lipofuscin , Microscopy, Electron, Transmission , Retinal Pigment Epithelium , Humans , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/ultrastructure , Retinal Pigment Epithelium/embryology , Adolescent , Autophagy/physiology , Child , Lipofuscin/metabolism , Exocytosis/physiology , Extracellular Space/metabolism , Gestational Age , Female , Male , Fetal Development/physiology , Mitochondria/metabolism , Mitochondria/ultrastructure , Cell Differentiation/physiology
9.
Am J Pathol ; 178(4): 1517-28, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21435441

ABSTRACT

This study was conducted to determine the perivascular cell responses to increased endothelial cell expression of insulin-like growth factor binding protein-3 (IGFBP-3) in mouse retina. The contribution of bone marrow cells in the IGFBP-3-mediated response was examined using green fluorescent protein-positive (GFP(+)) adult chimeric mice subjected to laser-induced retinal vessel occlusion injury. Intravitreal injection of an endothelial-specific IGFBP-3-expressing plasmid resulted in increased differentiation of GFP(+) hematopoietic stem cells (HSCs) into pericytes and astrocytes as determined by immunohistochemical analysis. Administration of IGFBP-3 plasmid to mouse pups that underwent the oxygen-induced retinopathy model resulted in increased pericyte ensheathment and reduced pericyte apoptosis in the developing retina. Increased IGFBP-3 expression reduced the number of activated microglial cells and decreased apoptosis of neuronal cells in the oxygen-induced retinopathy model. In summary, IGFBP-3 increased differentiation of GFP(+) HSCs into pericytes and astrocytes while increasing vascular ensheathment of pericytes and decreasing apoptosis of pericytes and retinal neurons. All of these cytoprotective effects exhibited by IGFBP-3 overexpression can result in a more stable retinal vascular bed. Thus, endothelial expression of IGFBP-3 may represent a physiologic response to injury and may represent a therapeutic strategy for the treatment of ischemic vascular eye diseases, such as diabetic retinopathy and retinopathy of prematurity.


Subject(s)
Apoptosis , Insulin-Like Growth Factor Binding Protein 3/metabolism , Ischemia/pathology , Microglia/metabolism , Neurons/metabolism , Pericytes/metabolism , Retina/injuries , Animals , Astrocytes/cytology , Cell Death , Cell Differentiation , Eye Diseases/pathology , Female , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/methods , Pericytes/cytology , Retina/metabolism
10.
Proc Natl Acad Sci U S A ; 106(36): 15489-94, 2009 Sep 08.
Article in English | MEDLINE | ID: mdl-19706893

ABSTRACT

The regulation of metal ion transport within neurons is critical for normal brain function. Of particular importance is the regulation of redox metals such as iron (Fe), where excess levels can contribute to oxidative stress and protein aggregation, leading to neuronal death. The divalent metal transporter 1 (DMT1) plays a central role in the regulation of Fe as well as other metals; hence, failure of DMT1 regulation is linked to human brain pathology. However, it remains unclear how DMT1 is regulated in the brain. Here, we show that DMT1 is regulated by Ndfip1 (Nedd4 family-interacting protein 1), an adaptor protein that recruits E3 ligases to ubiquitinate target proteins. Using human neurons we show the Ndfip1 is upregulated and binds to DMT1 in response to Fe and cobalt (Co) exposure. This interaction results in the ubiquitination and degradation of DMT1, resulting in reduced metal entry. Induction of Ndfip1 expression protects neurons from metal toxicity, and removal of Ndfip1 by shRNAi results in hypersensitivity to metals. We identify Nedd4-2 as an E3 ligase recruited by Ndfip1 for the ubiquitination of DMT1 within human neurons. Comparison of brains from Ndfip1(-/-) with Ndfip1(+/+) mice exposed to Fe reveals that Ndfip1(-/-) brains accumulate Fe within neurons. Together, this evidence suggests a critical role for Ndfip1 in regulating metal transport in human neurons.


Subject(s)
Carrier Proteins/metabolism , Cation Transport Proteins/metabolism , Cobalt/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Gene Expression Regulation/physiology , Iron/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Blotting, Western , Cobalt/toxicity , Gene Expression Regulation/drug effects , Humans , Immunohistochemistry , Immunoprecipitation , Ion Transport , Iron/toxicity , Mice , Mice, Knockout , Nedd4 Ubiquitin Protein Ligases , Neurons/drug effects , RNA Interference , Ubiquitination
11.
Exp Eye Res ; 92(5): 361-76, 2011 May.
Article in English | MEDLINE | ID: mdl-21354137

ABSTRACT

Human fetal eyes 8-40 weeks gestation (WG) were examined using markers to hematopoietic stem cells (HSC), vascular precursor cells (VPC), monocytes/macrophages and endothelial cells (EC). Electron microscopy and bromo-deoxyuridene labeling were undertaken to confirm the existence of solid vascular cords and to demonstrate vasculogenesis and angiogenesis in developing choroidal tissue. Our results demonstrated that the earliest incipient choroid consisted of vimentin(+) mesenchymal precursor cells which downregulated vimentin expression with maturation. Our observations lead us to conclude that these vimentin(-)/CD34(+)/CD44(+)/CD133(+) HSCs then differentiated into three distinct lineages: single isolated CD34(-)/CD39(+) VPCs that formed solid vascular cords which lumenized and became lined with CD34(+) vascular ECs; CD34(--+)/CD14(+)/CD68(+) monocytes that differentiated into tissue macrophages; and CD133(+)/CD34(--+)/α-smooth muscle actin(+) mural precursor cells that matured into smooth muscle cells and pericytes. Blood vessel formation occurred throughout the whole choroid simultaneously, indicative of in situ differentiation. Vasculogenesis, as evidenced by lumenization of solid vascular cords, was responsible for the formation of the entire choroidal area with angiogenesis, in all three layers of the choroid, only adding to vascular density. These results suggest that formation of the human choroid involves three processes: HSC differentiation, vasculogenesis and angiogenesis. Since vasculogenesis takes place independently of VEGF(165), further insights regarding the molecular mechanisms of vasculogenesis are required to better inform future treatments of choroidal neovascularization.


Subject(s)
Cell Differentiation/physiology , Choroid/blood supply , Choroid/embryology , Endothelium, Vascular/cytology , Hematopoietic Stem Cells/cytology , Neovascularization, Physiologic/physiology , Actins/metabolism , Antigens, CD/metabolism , Biomarkers/metabolism , Capillaries/cytology , Capillaries/metabolism , Cell Lineage , Endothelium, Vascular/metabolism , Gestational Age , Hematopoietic Stem Cells/metabolism , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Macrophages/cytology , Mesenchymal Stem Cells/cytology , Microscopy, Confocal , Microscopy, Electron , Vimentin/metabolism
12.
Circ Res ; 105(9): 897-905, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19762684

ABSTRACT

RATIONALE: Insulin-like growth factor binding protein (IGFBP)-3 modulates vascular development by regulating endothelial progenitor cell (EPC) behavior, specifically stimulating EPC cell migration. This study was undertaken to investigate the mechanism of IGFBP-3 effects on EPC function and how IGFBP-3 mediates cytoprotection following vascular injury. OBJECTIVE: To examine the mechanism of IGFBP-3-mediated repair following vascular injury. METHODS AND RESULTS: We used 2 complementary vascular injury models: laser occlusion of retinal vessels in adult green fluorescent protein (GFP) chimeric mice and oxygen-induced retinopathy in mouse pups. Intravitreal injection of IGFBP-3-expressing plasmid into lasered GFP chimeric mice stimulated homing of EPCs, whereas reversing ischemia induced increases in macrophage infiltration. IGFBP-3 also reduced the retinal ceramide/sphingomyelin ratio that was increased following laser injury. In the OIR model, IGFBP-3 prevented cell death of resident vascular endothelial cells and EPCs, while simultaneously increasing astrocytic ensheathment of vessels. For EPCs to orchestrate repair, these cells must migrate into ischemic tissue. This migratory ability is mediated, in part, by endogenous NO generation. Thus, we asked whether the migratory effects of IGFBP-3 were attributable to stimulation of NO generation. IGFBP-3 increased endothelial NO synthase expression in human EPCs leading to NO generation. IGFBP-3 exposure also led to the redistribution of vasodilator-stimulated phosphoprotein, an NO regulated protein critical for cell migration. IGFBP-3-mediated NO generation required high-density lipoprotein receptor activation and stimulation of phosphatidylinositol 3-kinase/Akt pathway. CONCLUSION: These studies support consideration of IGFBP-3 as a novel agent to restore the function of injured vasculature and restore NO generation.


Subject(s)
Cell Movement , Endothelial Cells/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Nitric Oxide/metabolism , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Retinopathy of Prematurity/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , Cell Adhesion Molecules/metabolism , Cell Death , Cell Proliferation , Cells, Cultured , Ceramides/metabolism , Cerebral Arteries/metabolism , Cerebral Arteries/physiopathology , Cytoprotection , Disease Models, Animal , Endothelial Cells/pathology , Female , Gene Transfer Techniques , Green Fluorescent Proteins/genetics , Humans , Infant, Newborn , Insulin-Like Growth Factor Binding Protein 3/genetics , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Retinal Neovascularization/pathology , Retinal Neovascularization/physiopathology , Retinal Vessels/pathology , Retinal Vessels/physiopathology , Retinopathy of Prematurity/pathology , Retinopathy of Prematurity/physiopathology , Scavenger Receptors, Class B/metabolism , Signal Transduction , Sphingomyelins/metabolism , Stem Cells/pathology , Vasodilation
13.
Glia ; 57(1): 39-53, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18661555

ABSTRACT

PURPOSE: To characterize the timing of differentiation, antigenic expression, morphology, proliferative potential, and apoptosis during astrocyte differentiation in the rat retina in vivo. METHODS: Whole mounts of rat retinae from embryonic day (E) 13 to postnatal day (P) 21 and adults were examined utilizing combinations of Pax2, GFAP, vimentin, S100, and GS lectin. These markers were also combined with BrdU and TUNEL to identify proliferation and apoptosis of cells of the astrocytic lineage. RESULTS: Three distinct stages of astrocytic differentiation were identified: (i) Pax2+/vimentin+/GFAP(-) astrocyte precursor cell (APC), (ii) Pax2+/vimentin+/GFAP+ immature perinatal astrocytes, and (iii) Pax2+/vimentin(-)/GFAP+ mature perinatal astrocytes. An earlier transient site of astrocyte generation was detected from E13 to E15 at the ventricular surface, but unlike the majority of retinal astrocytes that migrate into the retina starting at E15-E16, this ventricular source of retinal astrocytes were restricted to a small rim surrounding the optic nerve head. APCs and perinatal astrocytes were highly proliferative and migratory. Significant numbers of perinatal astrocytes were lost because of apoptosis, which was matched closely to the retraction of excess capillary segments during postnatal maturation of the retinal vasculature. CONCLUSIONS: This study provides evidence of a second site of astrocyte generation at the ventricular zone early in embryonic development of the mammalian retinae. APCs are present from E16 to E20 only during perinatal development and are a highly migratory and proliferative cell. As the retina is considered a part of the central nervous system (CNS), this is the first in vivo characterization of cells of the astrocytic lineage in mammalian CNS development.


Subject(s)
Apoptosis/physiology , Astrocytes/cytology , Cell Differentiation , Cell Proliferation , Retina/cytology , Stem Cells/cytology , Age Factors , Animals , Animals, Newborn , Astrocytes/physiology , Cell Differentiation/physiology , Cells, Cultured , Female , Pregnancy , Rats , Rats, Sprague-Dawley , Retina/embryology , Retina/growth & development , Stem Cells/physiology
14.
Sci Rep ; 9(1): 13464, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31530831

ABSTRACT

We apply novel analyses to images of superficial capillaries that are located near and around the optic disc of the human retina: the radial peri-papillary capillaries (RPCs). Due to their unique perfusion of the nerve fibre layer the RPCs are particularly significant for optic-neuropathies. The inputs to the analysis were z-stacks from 3D confocal fluorescence microscopy from 62 human retinas aged 9 to 84 years. Our aim was to find morphometric correlates of age. The retinas had no ophthalmic history. The analysis was undertaken in two stages: (1) converting the z-stacks to 3D tubular networks of vessels, and (2) characterizing the tubular networks using features derived from the Minkowski functionals (MFs). The MFs measure: the capillary volume, surface area, mean breadth, and Euler number. The mean breadth is related to tortuosity, wall shear stress and resistance to flow, and the Euler number is related to the density of loops (collaterals). Features derived from the surface area, mean breadth and Euler number were most related to age (all p ≤ 0.006). The results indicate the importance of pressure-equalizing loops and tortuosity as quantitative measures related to perfusion efficiency. The novel morphometric analysis could quantify disease-related accelerated aging and vessel malformation.


Subject(s)
Aging , Imaging, Three-Dimensional , Retinal Vessels/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Capillaries/physiology , Child , Humans , Microscopy, Confocal , Middle Aged , Retina , Young Adult
15.
Neural Regen Res ; 14(10): 1684-1694, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31169175

ABSTRACT

Neurogenesis is a persistent and essential feature of the adult mammalian hippocampus. Granular neurons generated from resident pools of stem or progenitor cells provide a mechanism for the formation and consolidation of new memories. Regulation of hippocampal neurogenesis is complex and multifaceted, and numerous signaling pathways converge to modulate cell proliferation, apoptosis, and clearance of cellular debris, as well as synaptic integration of newborn immature neurons. The expression of functional P2X7 receptors in the central nervous system has attracted much interest and the regulatory role of this purinergic receptor during adult neurogenesis has only recently begun to be explored. P2X7 receptors are exceptionally versatile: in their canonical role they act as adenosine triphosphate-gated calcium channels and facilitate calcium-signaling cascades exerting control over the cell via calcium-encoded sensory proteins and transcription factor activation. P2X7 also mediates transmembrane pore formation to regulate cytokine release and facilitate extracellular communication, and when persistently stimulated by high extracellular adenosine triphosphate levels large P2X7 pores form, which induce apoptotic cell death through cytosolic ion dysregulation. Lastly, as a scavenger receptor P2X7 directly facilitates phagocytosis of the cellular debris that arises during neurogenesis, as well as during some disease states. Understanding how P2X7 receptors regulate the physiology of stem and progenitor cells in the adult hippocampus is an important step towards developing useful therapeutic models for regenerative medicine. This review considers the relevant aspects of adult hippocampal neurogenesis and explores how P2X7 receptor activity may influence the molecular physiology of the hippocampus, and neural stem and progenitor cells.

16.
J Vis Exp ; (146)2019 04 03.
Article in English | MEDLINE | ID: mdl-31009002

ABSTRACT

Live-cell flow cytometry is increasingly used among cell biologists to quantify biological processes in a living cell culture. This protocol describes a method whereby live-cell flow cytometry is extended upon to analyze the multiple functions of P2X7 receptor activation in real-time. Using a time module installed on a flow cytometer, live-cell functionality can be assessed and plotted over a given time period to explore the kinetics of calcium influx, transmembrane pore formation, and phagocytosis. This simple method is advantageous as all three canonical functions of the P2X7 receptor can be assessed using one machine, and the gathered data plotted over time provides information on the entire live-cell population rather than single-cell recordings often obtained using technically challenging patch-clamp methods. Calcium influx experiments use a calcium indicator dye, while P2X7 pore formation assays rely on ethidium bromide being allowed to pass through the transmembrane pore formed upon high agonist concentrations. Yellow-green (YG) latex beads are utilized to measure phagocytosis. Specific agonists and antagonists are applied to investigate the effects of P2X7 receptor activity. Individually, these methods can be modified to provide quantitative data on any number of calcium channels and purinergic and scavenger receptors. Taken together, they highlight how the use of real-time live-cell flow cytometry is a rapid, cost-effective, reproducible, and quantifiable method to investigate P2X7 receptor function.


Subject(s)
Calcium/metabolism , Flow Cytometry/methods , Neural Stem Cells/cytology , Phagocytosis , Receptors, Purinergic P2X7/metabolism , Adenosine Triphosphate/pharmacology , Adult Stem Cells/metabolism , Animals , Ethidium/metabolism , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Patch-Clamp Techniques , Phagocytosis/drug effects
17.
Prog Retin Eye Res ; 62: 77-119, 2018 01.
Article in English | MEDLINE | ID: mdl-28958885

ABSTRACT

The population of infants at risk for retinopathy of prematurity (ROP) varies by world region; in countries with well developed neonatal intensive care services, the highest risk infants are those born at less than 28 weeks gestational age (GA) and less than 1 kg at birth, while, in regions where many aspects of neonatal intensive and ophthalmological care are not routinely available, more mature infants up to 2000 g at birth and 37 weeks GA are also at risk for severe ROP. Treatment options for both groups of patients include standard retinal laser photocoagulation or, more recently, intravitreal anti-VEGF drugs. In addition to detection and treatment of ROP, this review highlights new opportunities created by telemedicine, where screening and diagnosis of ROP in remote locations can be undertaken by non-ophthalmologists using digital fundus cameras. The ophthalmological care of the ROP infant is undertaken in the wider context of neonatal care and general wellbeing of the infant. Because of this context, this review takes a multi-disciplinary perspective with contributions from retinal vascular biologists, pediatric ophthalmologists, an epidemiologist and a neonatologist. This review highlights the latest insights regarding cellular and molecular mechanisms in the formation of the retinal vasculature in the human infant, pathogenesis of ROP, detection and treatment of severe ROP, the risks and benefits of anti-VEGF therapy, the identification of new therapies over the horizon, and the optimal neonatal care regimen for best ROP outcomes, and the benefits and pitfalls of telemedicine in the remote screening and diagnosis of ROP, all of which have the potential to improve ROP outcomes.


Subject(s)
Retinopathy of Prematurity , Angiogenesis Inhibitors/therapeutic use , Humans , Infant , Infant, Newborn , Laser Therapy/methods , Mass Screening , Retinal Vessels/embryology , Retinal Vessels/physiopathology , Retinopathy of Prematurity/diagnosis , Retinopathy of Prematurity/drug therapy , Retinopathy of Prematurity/physiopathology , Retinopathy of Prematurity/therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vitrectomy
18.
Invest Ophthalmol Vis Sci ; 48(3): 1368-79, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17325186

ABSTRACT

PURPOSE: Dilated and tortuous vessels (plus disease) in ROP is a grim prognostic indicator of visual outcome. The purpose of this study was to determine whether alterations in pericytes and smooth muscle cells (SMCs), are associated with the pathogenesis of ROP, including plus disease. METHODS: Kittens were exposed to either 4 (standard obliterative model) or 2 (modified model) days of hyperoxia, resulting in vaso-obliteration or localized vessel regression, respectively, and returned to room air. The modified model more closely resembles human ROP. Desmin and alpha-smooth muscle actin (SMA) immunohistochemistry and lectin labeling were used to label mural cells and vessels. The desmin ensheathment ratio (DER), a quantitative measure of vessel stability, was determined. RESULTS: In the neovasculature of the standard model and surviving vasculature of the modified model, radial arterioles and venules were dilated and SMCs attenuated. SMA expression on venules was decreased, and the difference in desmin expression normally observed between arterioles and venules was lost, indicating altered SMC differentiation. The DER was reduced in both ROP models, consistent with highly unstable vascular plexuses, receptive to angiogenic and vascular regression signals. CONCLUSIONS: The results provide compelling evidence of significant changes in arteriolar and venular SMCs in both experimental models of ROP. The delayed differentiation and apparent dedifferentiation of SMCs during the hypoxic phases would result in an impaired ability to regulate blood flow, contributing to the vasodilation and tortuosity, hallmarks of plus disease. Vessel tortuosity was seen only in the nonobliterative model, suggesting that tortuosity may be due to increased capillary resistance resulting from capillary closure.


Subject(s)
Disease Models, Animal , Muscle, Smooth, Vascular/pathology , Pericytes/pathology , Retinal Neovascularization/etiology , Retinal Vessels/ultrastructure , Retinopathy of Prematurity/etiology , Actins/metabolism , Animals , Animals, Newborn , Cats , Cell Differentiation , Desmin/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Hyperoxia/complications , Infant, Newborn , Muscle, Smooth, Vascular/metabolism , Pericytes/metabolism , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Vessels/metabolism , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology
19.
Invest Ophthalmol Vis Sci ; 58(12): 5043-5055, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28980000

ABSTRACT

Purpose: We investigated the relationship between inflammation, neuronal loss, and expression of indoleamine 2, 3-dioxygenase (IDO) and quinolinic acid (QUIN) in the retina of subjects with type 1 diabetes (T1D) and type 2 diabetes (T2D) and in the retina of rats with T1D. Methods: Retinas from T1D (n = 7), T2D (n = 13), and 20 age-matched nondiabetic human donors and from T1D (n = 3) and control rats (n = 3) were examined using immunohistochemistry for IDO, QUIN, cluster of differentiation 39 (CD39), ionized calcium-binding adaptor molecule (Iba-1, for macrophages and microglia), Vimentin (VIM; for Müller cells), neuronal nuclei (NeuN; for neurons), and UEA1 lectin (for blood vessels). Results: Based on morphologic criteria, CD39+/ionized calcium binding adaptor molecule 1(Iba-1+) resident microglia and CD39-/Iba-1+ bone marrow-derived macrophages were present at higher density in T1D (13% increase) and T2D (26% increase) human retinas when compared with controls. The density and brightness of IDO+ microglia were increased in both T1D and T2D human retinas. The intensity of QUIN+ expression on CD39+ microglia and VIM+ Müller cells was greatly increased in both human T1D and T2D retinas. T1D retinas showed a 63% loss of NeuN+ neurons and T2D retinas lost approximately 43% when compared with nondiabetic human retinas. Few QUIN+ microglia-like cells were seen in nondiabetic retinas, but the numbers increased 18-fold in T1D and 7-fold in T2D in the central retina. In T1D rat retinas, the density of IDO+ microglia increased 2.8-fold and brightness increased 2.1-fold when compared with controls. Conclusions: Our findings suggest that IDO and QUIN expression in the retinas of diabetic rats and humans could contribute to the neuronal degeneration that is characteristic of diabetic retinopathy.


Subject(s)
Biomarkers/metabolism , Diabetic Retinopathy/metabolism , Ependymoglial Cells/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Microglia/metabolism , Quinolinic Acid/metabolism , Retina/metabolism , Aged , Animals , Antigens, CD/metabolism , Antigens, Nuclear/metabolism , Apyrase/metabolism , Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetic Retinopathy/pathology , Ependymoglial Cells/pathology , Female , Fluorescent Antibody Technique, Indirect , Humans , Male , Microfilament Proteins/metabolism , Microglia/pathology , Microscopy, Confocal , Middle Aged , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Retina/pathology , Vimentin/metabolism
20.
J Neuroimmunol ; 173(1-2): 96-107, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16414123

ABSTRACT

Cerebral malaria (CM) kills more than 1 million children each year. Using a murine model of CM, we investigated the role of Fas-Fas ligand interactions in the neuropathogenesis of this disease. Lpr and Gld mice, deficient in Fas and Fas ligand, respectively, were protected from fatal CM, although they demonstrated some pathological features associated with CM in the wild type mouse. Fas-Fas ligand mRNA and protein expression were increased in the brain in mice with CM, and activated caspase-3-positive apoptotic astrocytes were observed. We suggest that Fas-mediated apoptosis of astrocytes is likely to be a critical factor in late-stage murine CM pathogenesis.


Subject(s)
Brain/pathology , Malaria, Cerebral/metabolism , Membrane Glycoproteins/metabolism , Tumor Necrosis Factors/metabolism , fas Receptor/metabolism , Animals , Apoptosis/physiology , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Disease Models, Animal , Fas Ligand Protein , Female , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Malaria, Cerebral/pathology , Mice , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL