Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Invest ; 133(4)2023 02 15.
Article in English | MEDLINE | ID: mdl-36787231

ABSTRACT

Pathological neovascularization in age-related macular degeneration (nvAMD) drives the principal cause of blindness in the elderly. While there is a robust genetic association between genes of innate immunity and AMD, genome-to-phenome relationships are low, suggesting a critical contribution of environmental triggers of disease. Possible insight comes from the observation that a past history of infection with pathogens such as Chlamydia pneumoniae, or other systemic inflammation, can predispose to nvAMD in later life. Using a mouse model of nvAMD with prior C. pneumoniae infection, endotoxin exposure, and genetic ablation of distinct immune cell populations, we demonstrated that peripheral infections elicited epigenetic reprogramming that led to a persistent memory state in retinal CX3CR1+ mononuclear phagocytes (MNPs). The immune imprinting persisted long after the initial inflammation had subsided and ultimately exacerbated choroidal neovascularization in a model of nvAMD. Single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) identified activating transcription factor 3 (ATF3) as a central mediator of retina-resident MNP reprogramming following peripheral inflammation. ATF3 polarized MNPs toward a reparative phenotype biased toward production of proangiogenic factors in response to subsequent injury. Therefore, a past history of bacterial endotoxin-induced inflammation can lead to immunological reprograming within CNS-resident MNPs and aggravate pathological angiogenesis in the aging retina.


Subject(s)
Choroidal Neovascularization , Macular Degeneration , Humans , Microglia/pathology , Retina/pathology , Choroidal Neovascularization/genetics , Macular Degeneration/genetics , Macular Degeneration/pathology , Inflammation/pathology
2.
Science ; 379(6627): 45-62, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36603072

ABSTRACT

Age-related macular degeneration is a prevalent neuroinflammatory condition and a major cause of blindness driven by genetic and environmental factors such as obesity. In diseases of aging, modifiable factors can be compounded over the life span. We report that diet-induced obesity earlier in life triggers persistent reprogramming of the innate immune system, lasting long after normalization of metabolic abnormalities. Stearic acid, acting through Toll-like receptor 4 (TLR4), is sufficient to remodel chromatin landscapes and selectively enhance accessibility at binding sites for activator protein-1 (AP-1). Myeloid cells show less oxidative phosphorylation and shift to glycolysis, ultimately leading to proinflammatory cytokine transcription, aggravation of pathological retinal angiogenesis, and neuronal degeneration associated with loss of visual function. Thus, a past history of obesity reprograms mononuclear phagocytes and predisposes to neuroinflammation.


Subject(s)
Epigenetic Memory , Immunity, Innate , Macular Degeneration , Neuroinflammatory Diseases , Obesity , Animals , Mice , Cytokines/genetics , Immunity, Innate/genetics , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/immunology , Obesity/genetics , Phagocytes/immunology , Transcription, Genetic , Macular Degeneration/genetics , Macular Degeneration/immunology , Cellular Reprogramming/genetics , Toll-Like Receptor 4/genetics
3.
Cell Metab ; 33(4): 818-832.e7, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33548171

ABSTRACT

Attenuating pathological angiogenesis in diseases characterized by neovascularization such as diabetic retinopathy has transformed standards of care. Yet little is known about the molecular signatures discriminating physiological blood vessels from their diseased counterparts, leading to off-target effects of therapy. We demonstrate that in contrast to healthy blood vessels, pathological vessels engage pathways of cellular senescence. Senescent (p16INK4A-expressing) cells accumulate in retinas of patients with diabetic retinopathy and during peak destructive neovascularization in a mouse model of retinopathy. Using either genetic approaches that clear p16INK4A-expressing cells or small molecule inhibitors of the anti-apoptotic protein BCL-xL, we show that senolysis suppresses pathological angiogenesis. Single-cell analysis revealed that subsets of endothelial cells with senescence signatures and expressing Col1a1 are no longer detected in BCL-xL-inhibitor-treated retinas, yielding a retina conducive to physiological vascular repair. These findings provide mechanistic evidence supporting the development of BCL-xL inhibitors as potential treatments for neovascular retinal disease.


Subject(s)
Cellular Senescence , Retinal Diseases/pathology , bcl-X Protein/metabolism , Animals , Apoptosis/drug effects , Cellular Senescence/drug effects , Collagen Type I, alpha 1 Chain/metabolism , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Flavonols/chemistry , Flavonols/pharmacology , Flavonols/therapeutic use , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic , Retinal Diseases/drug therapy , Retinal Diseases/metabolism , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , bcl-X Protein/antagonists & inhibitors
4.
Science ; 369(6506)2020 08 21.
Article in English | MEDLINE | ID: mdl-32820093

ABSTRACT

In developed countries, the leading causes of blindness such as diabetic retinopathy are characterized by disorganized vasculature that can become fibrotic. Although many such pathological vessels often naturally regress and spare sight-threatening complications, the underlying mechanisms remain unknown. Here, we used orthogonal approaches in human patients with proliferative diabetic retinopathy and a mouse model of ischemic retinopathies to identify an unconventional role for neutrophils in vascular remodeling during late-stage sterile inflammation. Senescent vasculature released a secretome that attracted neutrophils and triggered the production of neutrophil extracellular traps (NETs). NETs ultimately cleared diseased endothelial cells and remodeled unhealthy vessels. Genetic or pharmacological inhibition of NETosis prevented the regression of senescent vessels and prolonged disease. Thus, clearance of senescent retinal blood vessels leads to reparative vascular remodeling.


Subject(s)
Aging/pathology , Diabetic Retinopathy/pathology , Extracellular Traps/immunology , Retinal Vessels/pathology , Animals , Cellular Senescence , Diabetic Retinopathy/immunology , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Retinal Vessels/immunology
5.
Proc Natl Acad Sci U S A ; 116(10): 4538-4547, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30787185

ABSTRACT

Diabetic macular edema is a major complication of diabetes resulting in loss of central vision. Although heightened vessel leakiness has been linked to glial and neuronal-derived factors, relatively little is known on the mechanisms by which mature endothelial cells exit from a quiescent state and compromise barrier function. Here we report that endothelial NOTCH1 signaling in mature diabetic retinas contributes to increased vascular permeability. By providing both human and mouse data, we show that NOTCH1 ligands JAGGED1 and DELTA LIKE-4 are up-regulated secondary to hyperglycemia and activate both canonical and rapid noncanonical NOTCH1 pathways that ultimately disrupt endothelial adherens junctions in diabetic retinas by causing dissociation of vascular endothelial-cadherin from ß-catenin. We further demonstrate that neutralization of NOTCH1 ligands prevents diabetes-induced retinal edema. Collectively, these results identify a fundamental process in diabetes-mediated vascular permeability and provide translational rational for targeting the NOTCH pathway (primarily JAGGED1) in conditions characterized by compromised vascular barrier function.


Subject(s)
Capillary Permeability , Diabetic Retinopathy/pathology , Receptor, Notch1/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Calcium-Binding Proteins/biosynthesis , Enzyme Activation , Hyperglycemia/metabolism , Jagged-1 Protein/biosynthesis , Mice , Nitric Oxide/biosynthesis , Retinal Vessels/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , src-Family Kinases/metabolism
6.
Sci Transl Med ; 8(362): 362ra144, 2016 10 26.
Article in English | MEDLINE | ID: mdl-27797960

ABSTRACT

Pathological angiogenesis is the hallmark of diseases such as cancer and retinopathies. Although tissue hypoxia and inflammation are recognized as central drivers of vessel growth, relatively little is known about the process that bridges the two. In a mouse model of ischemic retinopathy, we found that hypoxic regions of the retina showed only modest rates of apoptosis despite severely compromised metabolic supply. Using transcriptomic analysis and inducible loss-of-function genetics, we demonstrated that ischemic retinal cells instead engage the endoplasmic reticulum stress inositol-requiring enzyme 1α (IRE1α) pathway that, through its endoribonuclease activity, induces a state of senescence in which cells adopt a senescence-associated secretory phenotype (SASP). We also detected SASP-associated cytokines (plasminogen activator inhibitor 1, interleukin-6, interleukin-8, and vascular endothelial growth factor) in the vitreous humor of patients suffering from proliferative diabetic retinopathy. Therapeutic inhibition of the SASP through intravitreal delivery of metformin or interference with effectors of senescence (semaphorin 3A or IRE1α) in mice reduced destructive retinal neovascularization in vivo. We conclude that the SASP contributes to pathological vessel growth, with ischemic retinal cells becoming prematurely senescent and secreting inflammatory cytokines that drive paracrine senescence, exacerbate destructive angiogenesis, and hinder reparative vascular regeneration. Reversal of this process may be therapeutically beneficial.


Subject(s)
Cellular Senescence , Diabetic Retinopathy/blood , Neovascularization, Pathologic , Retinal Vessels/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Cytokines/metabolism , Diabetic Retinopathy/physiopathology , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Oxygen/chemistry , Phenotype , Protein Serine-Threonine Kinases/metabolism , Retina/pathology , Retinal Neovascularization , Vascular Endothelial Growth Factor A/metabolism , Vitrectomy
SELECTION OF CITATIONS
SEARCH DETAIL
...