Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
J Clin Med ; 13(18)2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39337030

ABSTRACT

Background/Objectives: The objective of this study was to determine the treatment effect of foselutoclax in neovascular age-related macular degeneration (AMD) by multifocal electroretinography (mfERG) and evaluate mfERG as a potential clinical endpoint in AMD studies. Methods: A total of five subjects were included in the study who had active choroidal neovascularization and a history of at least two anti-vascular endothelial growth factor (VEGF) injections in the last 6 months. Subjects received a 50 µL intravitreal injection of foselutoclax at the baseline visit and Weeks 4, 24, and 28 of the study period. Results: After foselutoclax treatment, the largest improvement in the mfERG N1-P1 response density occurred at Week 8 as three of five subjects achieved a ≥20% gain. In addition, three of five subjects demonstrated a BCVA improvement of ≥5 ETDRS letters over baseline at Weeks 4, 8, and 24. The mean change in BCVA demonstrated statistical significance in Weeks 4 and 8, showing increases of 5 (p = 0.02) and 6.2 (p = 0.02) letters, respectively. Conclusions: Foselutoclax treatment was shown to have the potential to recover outer retinal function as determined by mfERG and BCVA at approximately Week 8 of treatment.

2.
Nat Med ; 30(2): 443-454, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38321220

ABSTRACT

Compromised vascular endothelial barrier function is a salient feature of diabetic complications such as sight-threatening diabetic macular edema (DME). Current standards of care for DME manage aspects of the disease, but require frequent intravitreal administration and are poorly effective in large subsets of patients. Here we provide evidence that an elevated burden of senescent cells in the retina triggers cardinal features of DME pathology and conduct an initial test of senolytic therapy in patients with DME. In cell culture models, sustained hyperglycemia provoked cellular senescence in subsets of vascular endothelial cells displaying perturbed transendothelial junctions associated with poor barrier function and leading to micro-inflammation. Pharmacological elimination of senescent cells in a mouse model of DME reduces diabetes-induced retinal vascular leakage and preserves retinal function. We then conducted a phase 1 single ascending dose safety study of UBX1325 (foselutoclax), a senolytic small-molecule inhibitor of BCL-xL, in patients with advanced DME for whom anti-vascular endothelial growth factor therapy was no longer considered beneficial. The primary objective of assessment of safety and tolerability of UBX1325 was achieved. Collectively, our data suggest that therapeutic targeting of senescent cells in the diabetic retina with a BCL-xL inhibitor may provide a long-lasting, disease-modifying intervention for DME. This hypothesis will need to be verified in larger clinical trials. ClinicalTrials.gov identifier: NCT04537884 .


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Macular Edema , Animals , Mice , Humans , Macular Edema/drug therapy , Macular Edema/etiology , Diabetic Retinopathy/drug therapy , Angiogenesis Inhibitors/therapeutic use , Endothelial Cells , Senotherapeutics , Cellular Senescence
3.
J Neuroinflammation ; 20(1): 145, 2023 Jun 21.
Article in English | MEDLINE | ID: mdl-37344842

ABSTRACT

Cellular adaptation to low oxygen tension triggers primitive pathways that ensure proper cell function. Conditions of hypoxia and low glucose are characteristic of injured tissues and hence successive waves of inflammatory cells must be suited to function under low oxygen tension and metabolic stress. While Hypoxia-Inducible Factor (HIF)-1α has been shown to be essential for the inflammatory response of myeloid cells by regulating the metabolic switch to glycolysis, less is known about how HIF1α is triggered in inflammation. Here, we demonstrate that cells of the innate immune system require activity of the inositol-requiring enzyme 1α (IRE1α/XBP1) axis in order to initiate HIF1α-dependent production of cytokines such as IL1ß, IL6 and VEGF-A. Knockout of either HIF1α or IRE1α in myeloid cells ameliorates vascular phenotypes in a model of retinal pathological angiogenesis driven by sterile inflammation. Thus, pathways associated with ER stress, in partnership with HIF1α, may co-regulate immune adaptation to low oxygen.


Subject(s)
Endoribonucleases , Protein Serine-Threonine Kinases , Humans , Protein Serine-Threonine Kinases/genetics , Hypoxia , Oxygen/metabolism , Myeloid Cells/metabolism , Inflammation/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit
4.
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
5.
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
6.
Proc Natl Acad Sci U S A ; 120(1): e2209973120, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36574648

ABSTRACT

Obesity is a major risk factor for cancer. Conventional thought suggests that elevated adiposity predisposes to heightened inflammatory stress and potentiates tumor growth, yet underlying mechanisms remain ill-defined. Here, we show that tumors from patients with a body mass index >35 carry a high burden of senescent cells. In mouse syngeneic tumor models, we correlated a pronounced accretion of senescent cancer cells with poorly immunogenic tumors when mice were subjected to diet-induced obesity (DIO). Highly immunogenic tumors showed lesser senescence burden suggesting immune-mediated elimination of senescent cancer cells, likely targeted as a consequence of their senescence-associated secretory phenotype. Treatment with the senolytic BH3 mimetic small molecule inhibitor ABT-263 selectively stalled tumor growth in mice with DIO to rates comparable to regular diet-fed mice. Thus, consideration of body adiposity in the selection of cancer therapy may be a critical determinant for disease outcome in poorly immunogenic malignancies.


Subject(s)
Cellular Senescence , Neoplasms , Mice , Animals , Obesity/complications
7.
Cells ; 11(14)2022 07 13.
Article in English | MEDLINE | ID: mdl-35883628

ABSTRACT

The GPCR SUCNR1/GPR91 exerts proangiogenesis upon stimulation with the Krebs cycle metabolite succinate. GPCR signaling depends on the surrounding environment and intracellular localization through location bias. Here, we show by microscopy and by cell fractionation that in neurons, SUCNR1 resides at the endoplasmic reticulum (ER), while being fully functional, as shown by calcium release and the induction of the expression of the proangiogenic gene for VEGFA. ER localization was found to depend upon N-glycosylation, particularly at position N8; the nonglycosylated mutant receptor localizes at the plasma membrane shuttled by RAB11. This SUCNR1 glycosylation is physiologically regulated, so that during hypoxic conditions, SUCNR1 is deglycosylated and relocates to the plasma membrane. Downstream signal transduction of SUCNR1 was found to activate the prostaglandin synthesis pathway through direct interaction with COX-2 at the ER; pharmacologic antagonism of the PGE2 EP4 receptor (localized at the nucleus) was found to prevent VEGFA expression. Concordantly, restoring the expression of SUCNR1 in the retina of SUCNR1-null mice renormalized vascularization; this effect is markedly diminished after transfection of the plasma membrane-localized SUCNR1 N8A mutant, emphasizing that ER localization of the succinate receptor is necessary for proper vascularization. These findings uncover an unprecedented physiologic process where GPCR resides at the ER for signaling function.


Subject(s)
Receptors, G-Protein-Coupled , Succinic Acid , Animals , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Hypoxia , Mice , Receptors, G-Protein-Coupled/metabolism , Succinates , Succinic Acid/metabolism
8.
JCI Insight ; 7(6)2022 03 22.
Article in English | MEDLINE | ID: mdl-35167498

ABSTRACT

Dyslipidemia and autophagy have been implicated in the pathogenesis of blinding neovascular age-related macular degeneration (NV-AMD). VLDL receptor (VLDLR), expressed in photoreceptors with a high metabolic rate, facilitates the uptake of triglyceride-derived fatty acids. Since fatty acid uptake is reduced in Vldlr-/- tissues, more remain in circulation, and the retina is fuel deficient, driving the formation in mice of neovascular lesions reminiscent of retinal angiomatous proliferation (RAP), a subtype of NV-AMD. Nutrient scarcity and energy failure are classically mitigated by increasing autophagy. We found that excess circulating lipids restrained retinal autophagy, which contributed to pathological angiogenesis in the Vldlr-/- RAP model. Triglyceride-derived fatty acid sensed by free fatty acid receptor 1 (FFAR1) restricted autophagy and oxidative metabolism in photoreceptors. FFAR1 suppressed transcription factor EB (TFEB), a master regulator of autophagy and lipid metabolism. Reduced TFEB, in turn, decreased sirtuin-3 expression and mitochondrial respiration. Metabolomic signatures of mouse RAP-like retinas were consistent with a role in promoting angiogenesis. This signature was also found in human NV-AMD vitreous. Restoring photoreceptor autophagy in Vldlr-/- retinas, either pharmacologically or by deleting Ffar1, enhanced metabolic efficiency and suppressed pathological angiogenesis. Dysregulated autophagy by circulating lipids might therefore contribute to the energy failure of photoreceptors driving neovascular eye diseases, and FFAR1 may be a target for intervention.


Subject(s)
Macular Degeneration , Retinal Neovascularization , Animals , Autophagy , Cell Proliferation , Fatty Acids , Macular Degeneration/pathology , Mice , Neovascularization, Pathologic , Receptors, G-Protein-Coupled , Retinal Neovascularization/pathology , Triglycerides
9.
Prog Retin Eye Res ; 90: 101051, 2022 09.
Article in English | MEDLINE | ID: mdl-35165043

ABSTRACT

Diabetic retinal disease (DRD), the most common complication of diabetes and a leading cause of blindness in working age individuals, is now understood to be a form of sensory neuropathy or neurovascular degeneration. Current treatments are focused on advanced vision-threatening disease and a single molecular target, vascular endothelial growth factor, has an approved therapy. We trace the evolution of understanding of DRD pathogenesis, identify new approaches to clinical assessment, trials infrastructure and design, and target identification to accelerate selection and evaluation of new therapeutics that will speed development of potentially curative interventions. Critically, the "Restoring Vision Moonshot" framework will address gaps in knowledge to be filled to achieve the goal of restoring sight and preventing vision loss in persons with diabetes.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Diabetic Retinopathy/pathology , Humans , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Vision Disorders
10.
Cell Mol Life Sci ; 79(1): 37, 2021 Dec 31.
Article in English | MEDLINE | ID: mdl-34971428

ABSTRACT

The roles of nitric oxide (NO) and endothelial NO synthase (eNOS) in the regulation of angiogenesis are well documented. However, the involvement of eNOS in the sprouting of endothelial tip-cells at the vascular front during sprouting angiogenesis remains poorly defined. In this study, we show that downregulation of eNOS markedly inhibits VEGF-stimulated migration of endothelial cells but increases their polarization, as evidenced by the reorientation of the Golgi in migrating monolayers and by the fewer filopodia on tip cells at ends of sprouts in endothelial cell spheroids. The effect of eNOS inhibition on EC polarization was prevented in Par3-depleted cells. Importantly, downregulation of eNOS increased the expression of polarity genes, such as PARD3B, PARD6A, PARD6B, PKCΖ, TJP3, and CRB1 in endothelial cells. In retinas of eNOS knockout mice, vascular development is retarded with decreased vessel density and vascular branching. Furthermore, tip cells at the extremities of the vascular front have a marked reduction in the number of filopodia per cell and are more oriented. In a model of oxygen-induced retinopathy (OIR), eNOS deficient mice are protected during the initial vaso-obliterative phase, have reduced pathological neovascularization, and retinal endothelial tip cells have fewer filopodia. Single-cell RNA sequencing of endothelial cells from OIR retinas revealed enrichment of genes related to cell polarity in the endothelial tip-cell subtype of eNOS deficient mice. These results indicate that inhibition of eNOS alters the polarity program of endothelial cells, which increases cell polarization, regulates sprouting angiogenesis and normalizes pathological neovascularization during retinopathy.


Subject(s)
Neovascularization, Pathologic , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/physiology , Retina/metabolism , Retinal Neovascularization , Retinal Vessels , Animals , Cattle , Cell Line , Cell Movement , Cell Polarity , Endothelial Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Retina/cytology , Retina/pathology , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Vessels/cytology , Retinal Vessels/pathology
11.
Nat Commun ; 12(1): 6984, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34848715

ABSTRACT

Eukaryotic cells have evolved highly orchestrated protein catabolic machineries responsible for the timely and selective disposal of proteins and organelles, thereby ensuring amino acid recycling. However, how protein degradation is coordinated with amino acid supply and protein synthesis has remained largely elusive. Here we show that the mammalian proteasome undergoes liquid-liquid phase separation in the nucleus upon amino acid deprivation. We termed these proteasome condensates SIPAN (Starvation-Induced Proteasome Assemblies in the Nucleus) and show that these are a common response of mammalian cells to amino acid deprivation. SIPAN undergo fusion events, rapidly exchange proteasome particles with the surrounding milieu and quickly dissolve following amino acid replenishment. We further show that: (i) SIPAN contain K48-conjugated ubiquitin, (ii) proteasome inhibition accelerates SIPAN formation, (iii) deubiquitinase inhibition prevents SIPAN resolution and (iv) RAD23B proteasome shuttling factor is required for SIPAN formation. Finally, SIPAN formation is associated with decreased cell survival and p53-mediated apoptosis, which might contribute to tissue fitness in diverse pathophysiological conditions.


Subject(s)
Amino Acids/metabolism , Apoptosis/physiology , Cell Nucleus/metabolism , Proteasome Endopeptidase Complex/metabolism , Starvation , Animals , Autoantigens , Cell Line, Tumor , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Eukaryotic Cells , Exercise , Fibroblasts , Humans , Mice , Nutrients , Protein Biosynthesis , Proteolysis , Stress, Physiological , Ubiquitin
12.
Sci Rep ; 11(1): 15767, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344941

ABSTRACT

The beneficial effects of brown adipose tissue (BAT) on obesity and associated metabolic diseases are mediated through its capacity to dissipate energy as heat. While immune cells, such as tissue-resident macrophages, are known to influence adipose tissue homeostasis, relatively little is known about their contribution to BAT function. Here we report that neuropilin-1 (NRP1), a multiligand single-pass transmembrane receptor, is highly expressed in BAT-resident macrophages. During diet-induced obesity (DIO), myeloid-resident NRP1 influences interscapular BAT mass, and consequently vascular morphology, innervation density and ultimately core body temperature during cold exposure. Thus, NRP1-expressing myeloid cells contribute to the BAT homeostasis and potentially its thermogenic function in DIO.


Subject(s)
Adipose Tissue, Brown/physiology , Homeostasis , Myeloid Cells/metabolism , Neuropilin-1/physiology , Obesity/prevention & control , Thermogenesis , Animals , Diet/adverse effects , Energy Metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/metabolism , Obesity/pathology
14.
EMBO Mol Med ; 13(5): e11754, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33876574

ABSTRACT

Age-related macular degeneration (AMD) in its various forms is a leading cause of blindness in industrialized countries. Here, we provide evidence that ligands for neuropilin-1 (NRP1), such as Semaphorin 3A and VEGF-A, are elevated in the vitreous of patients with AMD at times of active choroidal neovascularization (CNV). We further demonstrate that NRP1-expressing myeloid cells promote and maintain CNV. Expression of NRP1 on cells of myeloid lineage is critical for mitigating production of inflammatory factors such as IL6 and IL1ß. Therapeutically trapping ligands of NRP1 with an NRP1-derived trap reduces CNV. Collectively, our findings identify a role for NRP1-expressing myeloid cells in promoting pathological angiogenesis during CNV and introduce a therapeutic approach to counter neovascular AMD.


Subject(s)
Choroidal Neovascularization , Wet Macular Degeneration , Angiogenesis Inhibitors , Humans , Inflammation , Neuropilin-1/genetics , Vascular Endothelial Growth Factor A , Visual Acuity
15.
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
16.
Aging (Albany NY) ; 12(24): 24836-24852, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33361521

ABSTRACT

MicroRNAs are small non-coding RNAs that post-transcriptionally regulate gene expression. We recently demonstrated that levels of miR-106b were significantly decreased in the vitreous and plasma of patients with neovascular age-related macular degeneration (AMD). Here we show that expression of the miR-106b-25 cluster is negatively regulated by the unfolded protein response pathway of protein kinase RNA-like ER kinase (PERK) in a mouse model of neovascular AMD. A reduction in levels of miR-106b triggers vascular growth both in vivo and in vitro by inducing production of pro-angiogenic factors. We demonstrate that therapeutic delivery of miR-106b to the retina with lentiviral vectors protects against aberrant retinal angiogenesis in two distinct mouse models of pathological retinal neovascularization. Results from this study suggest that miRNAs such as miR-106b have the potential to be used as multitarget therapeutics for conditions characterized by pathological retinal angiogenesis.


Subject(s)
Choroidal Neovascularization/genetics , Macular Degeneration/genetics , MicroRNAs/genetics , Retinal Neovascularization/genetics , Animals , Cell Line , Cell Movement/genetics , Choroidal Neovascularization/pathology , Diabetic Retinopathy , Disease Models, Animal , Endoplasmic Reticulum Stress/genetics , Endothelial Cells , Eye Burns , Humans , Laser Therapy , Macular Degeneration/pathology , Mice , Oxygen/toxicity , Retinal Neovascularization/pathology , Retinopathy of Prematurity , Unfolded Protein Response/genetics , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
17.
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
18.
Immunity ; 53(2): 429-441.e8, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32814029

ABSTRACT

A minor haplotype of the 10q26 locus conveys the strongest genetic risk for age-related macular degeneration (AMD). Here, we examined the mechanisms underlying this susceptibility. We found that monocytes from homozygous carriers of the 10q26 AMD-risk haplotype expressed high amounts of the serine peptidase HTRA1, and HTRA1 located to mononuclear phagocytes (MPs) in eyes of non-carriers with AMD. HTRA1 induced the persistence of monocytes in the subretinal space and exacerbated pathogenic inflammation by hydrolyzing thrombospondin 1 (TSP1), which separated the two CD47-binding sites within TSP1 that are necessary for efficient CD47 activation. This HTRA1-induced inhibition of CD47 signaling induced the expression of pro-inflammatory osteopontin (OPN). OPN expression increased in early monocyte-derived macrophages in 10q26 risk carriers. In models of subretinal inflammation and AMD, OPN deletion or pharmacological inhibition reversed HTRA1-induced pathogenic MP persistence. Our findings argue for the therapeutic potential of CD47 agonists and OPN inhibitors for the treatment of AMD.


Subject(s)
CD47 Antigen/metabolism , Chromosomes, Human, Pair 10/genetics , High-Temperature Requirement A Serine Peptidase 1/metabolism , Macular Degeneration/genetics , Osteopontin/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Binding Sites/physiology , COS Cells , Cell Line , Chlorocebus aethiops , Eye/pathology , Genetic Predisposition to Disease/genetics , High-Temperature Requirement A Serine Peptidase 1/genetics , Humans , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Signal Transduction/genetics
19.
Br J Pharmacol ; 177(9): 1949-1966, 2020 05.
Article in English | MEDLINE | ID: mdl-31883121

ABSTRACT

BACKGROUND AND PURPOSE: Age-related macular degeneration (AMD) is a complex neurodegenerative disease treated by anti-VEGF intravitreal injections. As inflammation is potentially involved in retinal degeneration, the pro-inflammatory kallikrein-kinin system is a possible alternative pharmacological target. Here, we investigated the effects of anti-VEGF and anti-B1 receptor treatments on the inflammatory mechanisms in a rat model of choroidal neovascularization (CNV). EXPERIMENTAL APPROACH: Immediately after laser-induced CNV, Long-Evans rats were treated by eye-drop application of a B1 receptor antagonist (R-954) or by intravitreal injection of B1 receptor siRNA or anti-VEGF antibodies. Effects of treatments on gene expression of inflammatory mediators, CNV lesion regression and integrity of the blood-retinal barrier was measured 10 days later in the retina. B1 receptor and VEGF-R2 cellular localization was assessed. KEY RESULTS: The three treatments significantly inhibited the CNV-induced retinal changes. Anti-VEGF and R-954 decreased CNV-induced up-regulation of B1 and B2 receptors, TNF-α, and ICAM-1. Anti-VEGF additionally reversed up-regulation of VEGF-A, VEGF-R2, HIF-1α, CCL2 and VCAM-1, whereas R-954 inhibited gene expression of IL-1ß and COX-2. Enhanced retinal vascular permeability was abolished by anti-VEGF and reduced by R-954 and B1 receptor siRNA treatments. Leukocyte adhesion was impaired by anti-VEGF and B1 receptor inhibition. B1 receptors were found on astrocytes and endothelial cells. CONCLUSION AND IMPLICATIONS: B1 receptor and VEGF pathways were both involved in retinal inflammation and damage in laser-induced CNV. The non-invasive, self-administration of B1 receptor antagonists on the surface of the cornea by eye drops might be an important asset for the treatment of AMD.


Subject(s)
Choroidal Neovascularization , Neurodegenerative Diseases , Animals , Choroidal Neovascularization/drug therapy , Disease Models, Animal , Endothelial Cells , Inflammation/drug therapy , Kinins , Lasers , RNA, Messenger , Rats , Rats, Long-Evans , Retina
20.
Angiogenesis ; 23(2): 145-157, 2020 05.
Article in English | MEDLINE | ID: mdl-31598898

ABSTRACT

The Dll4-Notch-signaling pathway regulates capillary sprouting via the specification of endothelial tip cells. While VEGF is a potent inducer of Dll4 expression, the intracellular mediators that stimulate its expression remain poorly defined. The protein tyrosine phosphatase PTPRJ/DEP-1 is required for angiogenesis in normal or pathological contexts through its modulation of VEGF signaling. Here, we show that in DEP-1 KO mice, retinas at post-natal day 5 show enlarged blood vessels, as well as an increased number of tip cells and vessel branching points at the migrating front of the vascular plexus. Consistent with these observations, the proliferation of endothelial cells is increased in the retinas of DEP-1 KO mice, as revealed by phospho-histone H3 staining, and increased phosphorylation of ERK1/2 in HUVECs transfected with DEP-1 siRNA. The expression of Dll4 was decreased in retinas of DEP-1 KO mice and was associated with decreased Notch activation. Mechanistically, reduced Dll4 expression in the absence of DEP-1 was correlated with the inhibition of the Src/Akt/ß-Catenin-signaling pathway in HUVECs. Conversely, overexpression of WT DEP-1 in cultured endothelial cells, but not of mutants unable to activate Src-dependent signaling, promoted Dll4 expression. Inhibition of Src, Akt, and ß-catenin transcriptional activity, leading to the inhibition of Dll4 expression, further suggested that their activation through a DEP-1-dependent pathway was required to promote Dll4 expression in VEGF-stimulated endothelial cells. Altogether, these data demonstrate that DEP-1, via Akt and ß-catenin, is a significant promoter of the VEGF-induced Dll4-Notch pathway, and can contribute to the regulation of the tip and stalk cell phenotypes of endothelial cells.


Subject(s)
Endothelial Cells , Neovascularization, Physiologic , Receptor-Like Protein Tyrosine Phosphatases, Class 3 , Receptors, Notch , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Endothelial Cells/metabolism , Mice , Neovascularization, Physiologic/genetics , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL