Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancer Res ; 77(20): 5664-5675, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28790065

ABSTRACT

Maturation of human natural killer (NK) cells as defined by accumulation of cell-surface expression of CD57 is associated with increased cytotoxic character and TNF and IFNγ production upon target-cell recognition. Notably, multiple studies point to a unique role for CD57+ NK cells in cancer immunosurveillance, yet there is scant information about how they mature. In this study, we show that pharmacologic inhibition of GSK3 kinase in peripheral blood NK cells expanded ex vivo with IL15 greatly enhances CD57 upregulation and late-stage maturation. GSK3 inhibition elevated the expression of several transcription factors associated with late-stage NK-cell maturation including T-BET, ZEB2, and BLIMP-1 without affecting viability or proliferation. When exposed to human cancer cells, NK cell expanded ex vivo in the presence of a GSK3 inhibitor exhibited significantly higher production of TNF and IFNγ, elevated natural cytotoxicity, and increased antibody-dependent cellular cytotoxicity. In an established mouse xenograft model of ovarian cancer, adoptive transfer of NK cells conditioned in the same way also displayed more robust and durable tumor control. Our findings show how GSK3 kinase inhibition can greatly enhance the mature character of NK cells most desired for effective cancer immunotherapy. Cancer Res; 77(20); 5664-75. ©2017 AACR.


Subject(s)
Glycogen Synthase Kinase 3/antagonists & inhibitors , Immunotherapy, Adoptive/methods , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , A549 Cells , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Female , Glycogen Synthase Kinase 3/immunology , Humans , Interleukin-15/pharmacology , K562 Cells , Killer Cells, Natural/enzymology , Mice , Mice, Inbred NOD , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Pyridines/pharmacology , Pyrimidines/pharmacology , Xenograft Model Antitumor Assays
2.
J Vis Exp ; (116)2016 10 18.
Article in English | MEDLINE | ID: mdl-27805590

ABSTRACT

Microglia are the tissue resident macrophages of the central nervous system (CNS) and they perform a variety of functions that support CNS homeostasis, including phagocytosis of damaged synapses or cells, debris, and/or invading pathogens. Impaired phagocytic function has been implicated in the pathogenesis of diseases such as Alzheimer's and age-related macular degeneration, where amyloid-ß plaque and drusen accumulate, respectively. Despite its importance, microglial phagocytosis has been challenging to assess in vivo. Here, we describe a simple, yet robust, technique for precisely monitoring and quantifying the in vivo phagocytic potential of retinal microglia. Previous methods have relied on immunohistochemical staining and imaging techniques. Our method uses flow cytometry to measure microglial uptake of fluorescently labeled particles after intravitreal delivery to the eye in live rodents. This method replaces conventional practices that involve laborious tissue sectioning, immunostaining, and imaging, allowing for more precise quantification of microglia phagocytic function in just under six hours. This procedure can also be adapted to test how various compounds alter microglial phagocytosis in physiological settings. While this technique was developed in the eye, its use is not limited to vision research.


Subject(s)
Flow Cytometry/methods , Neuroglia , Phagocytes , Retina , Humans , Microglia , Phagocytosis
3.
J Clin Invest ; 125(6): 2335-46, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25915585

ABSTRACT

Functional interactions between neurons, vasculature, and glia within neurovascular units are critical for maintenance of the retina and other CNS tissues. For example, the architecture of the neurosensory retina is a highly organized structure with alternating layers of neurons and blood vessels that match the metabolic demand of neuronal activity with an appropriate supply of oxygen within perfused blood. Here, using murine genetic models and cell ablation strategies, we have demonstrated that a subset of retinal interneurons, the amacrine and horizontal cells, form neurovascular units with capillaries in 2 of the 3 retinal vascular plexuses. Moreover, we determined that these cells are required for generating and maintaining the intraretinal vasculature through precise regulation of hypoxia-inducible and proangiogenic factors, and that amacrine and horizontal cell dysfunction induces alterations to the intraretinal vasculature and substantial visual deficits. These findings demonstrate that specific retinal interneurons and the intraretinal vasculature are highly interdependent, and loss of either or both elicits profound effects on photoreceptor survival and function.


Subject(s)
Amacrine Cells/metabolism , Capillaries/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retinal Horizontal Cells/metabolism , Retinal Vessels/metabolism , Vision, Ocular/physiology , Amacrine Cells/cytology , Animals , Capillaries/cytology , Mice , Mice, Transgenic , Photoreceptor Cells, Vertebrate/cytology , Retinal Horizontal Cells/cytology , Retinal Vessels/cytology
4.
J Clin Invest ; 123(11): 4900-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24084735

ABSTRACT

Vascular networks develop from a growing vascular front that responds to VEGF and other guidance cues. Angiogenesis is required for normal tissue function, but, under conditions of stress, inappropriate vascularization can lead to disease. Therefore, inhibition of angiogenic sprouting may prevent neovascularization in patients with blinding neovascular eye diseases, including macular degeneration. VEGF antagonists have therapeutic benefits but also can elicit off-target effects. Here, we found that the Ras pathway, which functions downstream of a wide range of cytokines including VEGF, is active in the growing vascular front of developing and pathological vascular networks. The endogenous Ras inhibitor p120RasGAP was expressed predominately in quiescent VEGF-insensitive endothelial cells and was ectopically downregulated in multiple neovascular models. MicroRNA-132 negatively regulated p120RasGAP expression. Experimental delivery of α-miR-132 to developing mouse eyes disrupted tip cell Ras activity and prevented angiogenic sprouting. This strategy prevented ocular neovascularization in multiple rodent models even more potently than the VEGF antagonist, VEGF-trap. Targeting microRNA-132 as a therapeutic strategy may prove useful for treating multiple neovascular diseases of the eye and for preventing vision loss regardless of the neovascular stimulus.


Subject(s)
Endothelial Cells/metabolism , Endothelial Cells/pathology , Neovascularization, Pathologic/prevention & control , ras Proteins/antagonists & inhibitors , Animals , Disease Models, Animal , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Neovascularization/prevention & control , Retinal Vessels/growth & development , Retinal Vessels/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , p120 GTPase Activating Protein/metabolism , ras Proteins/metabolism
5.
Invest Ophthalmol Vis Sci ; 53(10): 6282-90, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22871841

ABSTRACT

PURPOSE: Retinal pigment epithelium (RPE) autologous grafts can be readily derived from induced pluripotent stem (iPS) cells. It is critical to stringently characterize iPS-RPE using standardized and quantifiable methods to be confident that they are safe and adequate replacements for diseased RPE before utilizing them in clinical settings. One important and required function is that the iPS-RPE phagocytose photoreceptor outer segments (POS). METHODS: We developed a flow cytometry-based assay to monitor binding and internalization of FITC labeled POS by ARPE-19, human fetal RPE (hfRPE), and two types of iPS-RPE. Expression and density of α(v)ß5 integrin, CD36, and MerTK receptors, which are required for phagocytosis, were compared. RESULTS: Trypsinization of treated RPE cells results in the release of bound POS. The number of freed POS, the percentage of cells that internalized POS, the brightness of the FITC signal from the cells, and the surface density of the phagocytosis receptors on single RPE cells were measured using flow cytometry. These assays reveal that receptor density is dynamic during differentiation and this can affect the binding and internalization dynamics of the RPE cells. Highly differentiated iPS-RPE phagocytose POS more efficiently than hfRPE. CONCLUSIONS: Caution should be exercised to not use RPE grafts until demonstrating that they are fully functional. The density of the phagocytosis receptors is dynamic and may be used as a predictor for how well the iPS-RPE cells will function in vivo. The phagocytosis dynamics observed between iPS-RPE and primary RPE is very encouraging and adds to mounting evidence that iPS-RPE may be a viable replacement for dysfunctional or dying RPE in human patients.


Subject(s)
Flow Cytometry/methods , Induced Pluripotent Stem Cells/cytology , Phagocytosis/physiology , Retinal Photoreceptor Cell Outer Segment/physiology , Animals , Eye Proteins/metabolism , Humans , Receptors, Immunologic/metabolism , Swine
6.
Sci Rep ; 1: 76, 2011.
Article in English | MEDLINE | ID: mdl-22355595

ABSTRACT

Diabetic retinopathy is the leading cause of visual loss in individuals under the age of 55. Umbilical cord blood (UCB)-derived myeloid progenitor cells have been shown to decrease neuronal damage associated with ischemia in the central nervous system. In this study we show that UCB-derived CD14(+) progenitor cells provide rescue effects in a mouse model of ischemic retinopathy by promoting physiological angiogenesis and reducing associated inflammation. We use confocal microscopy to trace the fate of injected human UCB-derived CD14(+) cells and PCR with species-specific probes to investigate their gene expression profile before and after injection. Metabolomic analysis measures changes induced by CD14(+) cells. Our results demonstrate that human cells differentiate in vivo into M2 macrophages and induce the polarization of resident M2 macrophages. This leads to stabilization of the ischemia-injured retinal vasculature by modulating the inflammatory response, reducing oxidative stress and apoptosis and promoting tissue repair.


Subject(s)
Disease Models, Animal , Ischemia/pathology , Macrophages/physiology , Retinal Diseases/pathology , Retinal Vessels/pathology , Animals , Cells, Cultured , Humans , Lipopolysaccharide Receptors/immunology , Macrophages/immunology , Mice , Microscopy, Confocal , Polymerase Chain Reaction
7.
Methods Enzymol ; 444: 115-58, 2008.
Article in English | MEDLINE | ID: mdl-19007663

ABSTRACT

During normal retinal vascular development, vascular endothelial cells proliferate and migrate through the extracellular matrix in response to a variety of cytokines, leading to the formation of new blood vessels in a highly ordered fashion. However, abnormal angiogenesis contributes to the vast majority of diseases that cause catastrophic loss of vision. During abnormal neovascularization of the iris, retina, or choroid, angiogenesis is unregulated and usually results in the formation of dysfunctional blood vessels. Multiple models of ocular angiogenesis exist which recapitulate particular aspects of both normal and pathological neovascularization. These experimental methods are useful for studying the mechanisms of normal developmental angiogenesis, as well as studying various aspects of pathological angiogenesis including ischemic retinopathies, vascular leak, and choroidal neovascularization. This chapter will outline several protocols used to study ocular angiogenesis, put the protocols into brief historical context, and describe some of the questions for which these protocols are commonly used.


Subject(s)
Eye/blood supply , Models, Biological , Neovascularization, Physiologic , Animals , Eye Diseases/chemically induced , Mice , Ophthalmologic Surgical Procedures , Oxygen/pharmacology
8.
Angiogenesis ; 10(2): 89-101, 2007.
Article in English | MEDLINE | ID: mdl-17372851

ABSTRACT

Nothing more dramatically captures the imagination of the visually impaired patient or the ophthalmologist treating them than the possibility of rebuilding a damaged retina or vasculature with "stem cells." Stem cells (SC) have been isolated from adult tissues and represent a pool of cells that may serve to facilitate rescue/repair of damaged tissue following injury or stress. We propose a new paradigm to "mature" otherwise immature neovasculature or, better yet, stabilize existing vasculature to hypoxic damage. This may be possible through the use of autologous bone marrow (BM) or cord blood derived hematopoietic SC that selectively target sites of neovascularization and gliosis where they provide vasculo- and neurotrophic effects. We have demonstrated that adult BM contains a population of endothelial and myeloid progenitor cells that can target activated astrocytes, a hallmark of many ocular diseases, and participate in normal developmental, or injury-induced, angiogenesis in the adult. Intravitreal injection of these cells from mice and humans can prevent retinal vascular degeneration ordinarily observed in mouse models of retinal degeneration; this vascular rescue correlates with functional neuronal rescue as well. The use of autologous adult BM derived SC grafts for the treatment of retinal vascular and degenerative diseases represents a novel conceptual approach that may make it possible to "mature" otherwise immature neovasculature, stabilize existing vasculature to hypoxic damage and/or rescue and protect retinal neurons from undergoing apoptosis. Such a therapeutic approach would obviate the need to employ destructive treatment modalities and would facilitate vascularization of ischemic and otherwise damaged retinal tissue.


Subject(s)
Hematopoietic Stem Cell Transplantation , Neovascularization, Physiologic , Retinal Diseases/physiopathology , Retinal Diseases/therapy , Retinal Vessels/physiology , Stem Cell Transplantation , Adult , Bone Marrow Cells/physiology , Cord Blood Stem Cell Transplantation , Endothelium, Vascular/cytology , Humans , Retinal Vessels/physiopathology , Safety
9.
J Clin Invest ; 116(12): 3266-76, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17111048

ABSTRACT

Vision loss associated with ischemic diseases such as retinopathy of prematurity and diabetic retinopathy are often due to retinal neovascularization. While significant progress has been made in the development of compounds useful for the treatment of abnormal vascular permeability and proliferation, such therapies do not address the underlying hypoxia that stimulates the observed vascular growth. Using a model of oxygen-induced retinopathy, we demonstrate that a population of adult BM-derived myeloid progenitor cells migrated to avascular regions of the retina, differentiated into microglia, and facilitated normalization of the vasculature. Myeloid-specific hypoxia-inducible factor 1alpha (HIF-1alpha) expression was required for this function, and we also demonstrate that endogenous microglia participated in retinal vascularization. These findings suggest what we believe to be a novel therapeutic approach for the treatment of ischemic retinopathies that promotes vascular repair rather than destruction.


Subject(s)
Cell Differentiation , Microglia/cytology , Myeloid Progenitor Cells/cytology , Retinal Diseases/metabolism , Animals , Bone Marrow/metabolism , Cells, Cultured , Disease Models, Animal , Electroretinography/methods , Female , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Immunohistochemistry , Ischemia/complications , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microglia/metabolism , Myeloid Progenitor Cells/physiology , Retina/metabolism , Retina/pathology , Retina/physiopathology , Retinal Diseases/etiology , Retinal Diseases/physiopathology , Retinal Neovascularization/etiology , Retinal Neovascularization/metabolism , Retinal Neovascularization/physiopathology , Wound Healing/physiology
10.
J Clin Invest ; 114(6): 765-74, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15372100

ABSTRACT

Inherited retinal degenerations afflict 1 in 3,500 individuals and are a heterogeneous group of diseases that result in profound vision loss, usually the result of retinal neuronal apoptosis. Atrophic changes in the retinal vasculature are also observed in many of these degenerations. While it is thought that this atrophy is secondary to diminished metabolic demand in the face of retinal degeneration, the precise relationship between the retinal neuronal and vascular degeneration is not clear. In this study we demonstrate that whenever a fraction of mouse or human adult bone marrow-derived stem cells (lineage-negative hematopoietic stem cells [Lin- HSCs]) containing endothelial precursors stabilizes and rescues retinal blood vessels that would ordinarily completely degenerate, a dramatic neurotrophic rescue effect is also observed. Retinal nuclear layers are preserved in 2 mouse models of retinal degeneration, rd1 and rd10, and detectable, albeit severely abnormal, electroretinogram recordings are observed in rescued mice at times when they are never observed in control-treated or untreated eyes. The normal mouse retina consists predominantly of rods, but the rescued cells after treatment with Lin- HSCs are nearly all cones. Microarray analysis of rescued retinas demonstrates significant upregulation of many antiapoptotic genes, including small heat shock proteins and transcription factors. These results suggest a new paradigm for thinking about the relationship between vasculature and associated retinal neuronal tissue as well as a potential treatment for delaying the progression of vision loss associated with retinal degeneration regardless of the underlying genetic defect.


Subject(s)
Retinal Degeneration/therapy , Retinal Vessels/physiopathology , Stem Cell Transplantation/methods , Adult , Animals , Bone Marrow Cells/cytology , Cell Nucleus/pathology , Disease Models, Animal , Electroretinography , Humans , Injections , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Retinal Degeneration/pathology , Retinal Vessels/pathology , Vitreous Body
11.
Blood ; 103(9): 3420-7, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14726407

ABSTRACT

Adult bone marrow contains a population of hematopoietic stem cells (HSCs) that can give rise to cells capable of targeting sites of neovascularization in the peripheral or retinal vasculature. However, relatively little is known about the mechanism of targeting of these cells to sites of neovascularization. We have analyzed subpopulations of HSCs for the expression of a variety of cell surface adhesion molecules and found that R-cadherin, a calcium-dependent cell-cell adhesion molecule important for normal retinal endothelial cell guidance, was preferentially expressed by functionally targeting HSCs. Preincubation of HSCs with function-blocking anti-R-cadherin antibodies or novel R-cadherin-specific peptide antagonists effectively prevented targeting of bone marrow-derived cells to the developing retinal vasculature in vivo. Whereas control-injected HSCs targeted to all 3 normal developing retinal vascular layers, blocking R-cadherin-mediated adhesion resulted in mistargeting of the HSCs to the normally avascular outer retina. Our results suggest that vascular targeting of bone marrow-derived HSCs is dependent on mechanisms similar to those used by endogenous retinal vascular endothelial cells. Thus, R-cadherin antagonists may be useful in the treatment of neovascular diseases in which circulating HSCs contribute to abnormal angiogenesis.


Subject(s)
Cadherins/physiology , Cell Movement , Hematopoietic Stem Cells/cytology , Neovascularization, Physiologic , Retina/growth & development , Animals , Bone Marrow Cells/cytology , Cadherins/analysis , Cadherins/drug effects , Cell Adhesion , Hematopoietic Stem Cells/chemistry , Mice , Mice, Transgenic , Peptides/pharmacology , Retina/cytology , Retinal Vessels
12.
Lymphat Res Biol ; 1(4): 291-9, 2003.
Article in English | MEDLINE | ID: mdl-15624557

ABSTRACT

Hemangiomas are benign endothelial tumors. Often referred to as hemangiomas of infancy (HOI), these tumors are the most common tumor of infancy. Most of these lesions proliferate rapidly in the first months of life, and subsequently slowly involute during early childhood without significant complications. However, they often develop on the head or neck, and may pose a significant cosmetic concern for families. In addition, a fraction of these tumors can grow explosively and ulcerate, bleed, or obstruct vision or airway structures. Current treatments for these tumors are associated with significant side effects, and our knowledge of the biology of hemangiomas is limited. The natural evolution of these lesions creates a unique opportunity to study the changes in gene expression that occur as the endothelium of these tumors proliferates and then subsequently regresses. Such information may also increase our understanding of the basic principals of angiogenesis in normal and abnormal tissue. We have performed large-scale genomic analysis of hemangioma gene expression using DNA microarrays. We recently identified insulin-like growth factor 2 as a potentially important regulator of hemangioma growth using this approach. However, little is known about the mechanisms involved in hemangioma involution. Here we explore the idea that hemangioma involution might be an immune-mediated process and present data to support this concept. We also demonstrate that proliferating hemangiomas express indoleamine 2,3 dioxygenase (IDO) and discuss a possible mechanism that accounts for the often slow regression of these lesions.


Subject(s)
Hemangioma/immunology , Hemangioma/pathology , Tryptophan Oxygenase/metabolism , CD3 Complex/biosynthesis , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Dioxygenases , Disease Progression , Hemangioma, Capillary , Humans , Immunoblotting , Immunohistochemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase , Infant, Newborn , Insulin-Like Growth Factor II/metabolism , Intercellular Adhesion Molecule-1/biosynthesis , Oligonucleotide Array Sequence Analysis , T-Lymphocytes/metabolism , Time Factors , Vascular Cell Adhesion Molecule-1/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...