Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Sci Immunol ; 6(66): eabi4493, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34860583

ABSTRACT

Detection of microbial products by multiprotein complexes known as inflammasomes is pivotal to host defense against pathogens. Nucleotide-binding domain leucine-rich repeat (NLR) CARD domain containing 4 (NLRC4) forms an inflammasome in response to bacterial products; this requires their detection by NLR family apoptosis inhibitory proteins (NAIPs), with which NLRC4 physically associates. However, the mechanisms underlying sterile NLRC4 inflammasome activation, which is implicated in chronic noninfectious diseases, remain unknown. Here, we report that endogenous short interspersed nuclear element (SINE) RNAs, which promote atrophic macular degeneration (AMD) and systemic lupus erythematosus (SLE), induce NLRC4 inflammasome activation independent of NAIPs. We identify DDX17, a DExD/H box RNA helicase, as the sensor of SINE RNAs that licenses assembly of an inflammasome comprising NLRC4, NLR pyrin domain­containing protein 3, and apoptosis-associated speck-like protein­containing CARD and induces caspase-1 activation and cytokine release. Inhibiting DDX17-mediated NLRC4 inflammasome activation decreased interleukin-18 release in peripheral blood mononuclear cells of patients with SLE and prevented retinal degeneration in an animal model of AMD. Our findings uncover a previously unrecognized noncanonical NLRC4 inflammasome activated by endogenous retrotransposons and provide potential therapeutic targets for SINE RNA­driven diseases.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , DEAD-box RNA Helicases/immunology , Inflammasomes/immunology , RNA/immunology , Retroelements/immunology , Animals , Apoptosis Regulatory Proteins/deficiency , Calcium-Binding Proteins/deficiency , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Article in English | MEDLINE | ID: mdl-34620711

ABSTRACT

The atrophic form of age-related macular degeneration (dry AMD) affects nearly 200 million people worldwide. There is no Food and Drug Administration (FDA)-approved therapy for this disease, which is the leading cause of irreversible blindness among people over 50 y of age. Vision loss in dry AMD results from degeneration of the retinal pigmented epithelium (RPE). RPE cell death is driven in part by accumulation of Alu RNAs, which are noncoding transcripts of a human retrotransposon. Alu RNA induces RPE degeneration by activating the NLRP3-ASC inflammasome. We report that fluoxetine, an FDA-approved drug for treating clinical depression, binds NLRP3 in silico, in vitro, and in vivo and inhibits activation of the NLRP3-ASC inflammasome and inflammatory cytokine release in RPE cells and macrophages, two critical cell types in dry AMD. We also demonstrate that fluoxetine, unlike several other antidepressant drugs, reduces Alu RNA-induced RPE degeneration in mice. Finally, by analyzing two health insurance databases comprising more than 100 million Americans, we report a reduced hazard of developing dry AMD among patients with depression who were treated with fluoxetine. Collectively, these studies identify fluoxetine as a potential drug-repurposing candidate for dry AMD.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Drug Repositioning/methods , Fluoxetine/pharmacology , Macular Degeneration/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Retinal Pigment Epithelium/drug effects , Alu Elements/genetics , Animals , Blindness/pathology , Blindness/prevention & control , Cell Line , Cytokines/metabolism , Depression/drug therapy , Disease Models, Animal , Inflammasomes/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , RNA/genetics , Retina/pathology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/pathology
3.
Sci Adv ; 7(40): eabj3658, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34586848

ABSTRACT

Long interspersed nuclear element-1 (L1)­mediated reverse transcription (RT) of Alu RNA into cytoplasmic Alu complementary DNA (cDNA) has been implicated in retinal pigmented epithelium (RPE) degeneration. The mechanism of Alu cDNA­induced cytotoxicity and its relevance to human disease are unknown. Here we report that Alu cDNA is highly enriched in the RPE of human eyes with geographic atrophy, an untreatable form of age-related macular degeneration. We demonstrate that the DNA sensor cGAS engages Alu cDNA to induce cytosolic mitochondrial DNA escape, which amplifies cGAS activation, triggering RPE degeneration via the inflammasome. The L1-extinct rice rat was resistant to Alu RNA­induced Alu cDNA synthesis and RPE degeneration, which were enabled upon L1-RT overexpression. Nucleoside RT inhibitors (NRTIs), which inhibit both L1-RT and inflammasome activity, and NRTI derivatives (Kamuvudines) that inhibit inflammasome, but not RT, both block Alu cDNA toxicity, identifying inflammasome activation as the terminal effector of RPE degeneration.

4.
Signal Transduct Target Ther ; 6(1): 149, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33850097

ABSTRACT

Nonfibrillar amyloid-ß oligomers (AßOs) are a major component of drusen, the sub-retinal pigmented epithelium (RPE) extracellular deposits characteristic of age-related macular degeneration (AMD), a common cause of global blindness. We report that AßOs induce RPE degeneration, a clinical hallmark of geographic atrophy (GA), a vision-threatening late stage of AMD that is currently untreatable. We demonstrate that AßOs induce activation of the NLRP3 inflammasome in the mouse RPE in vivo and that RPE expression of the purinergic ATP receptor P2RX7, an upstream mediator of NLRP3 inflammasome activation, is required for AßO-induced RPE degeneration. Two classes of small molecule inflammasome inhibitors-nucleoside reverse transcriptase inhibitors (NRTIs) and their antiretrovirally inert modified analog Kamuvudines-both inhibit AßOs-induced RPE degeneration. These findings crystallize the importance of P2RX7 and NLRP3 in a disease-relevant model of AMD and identify inflammasome inhibitors as potential treatments for GA.


Subject(s)
Amyloid beta-Peptides/metabolism , Macular Degeneration/drug therapy , Retinal Pigment Epithelium/metabolism , Reverse Transcriptase Inhibitors/pharmacology , Amyloid beta-Peptides/genetics , Animals , Disease Models, Animal , Humans , Macular Degeneration/genetics , Macular Degeneration/metabolism , Male , Mice , Mice, Knockout
5.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Article in English | MEDLINE | ID: mdl-33526699

ABSTRACT

Alu retroelements propagate via retrotransposition by hijacking long interspersed nuclear element-1 (L1) reverse transcriptase (RT) and endonuclease activities. Reverse transcription of Alu RNA into complementary DNA (cDNA) is presumed to occur exclusively in the nucleus at the genomic integration site. Whether Alu cDNA is synthesized independently of genomic integration is unknown. Alu RNA promotes retinal pigmented epithelium (RPE) death in geographic atrophy, an untreatable type of age-related macular degeneration. We report that Alu RNA-induced RPE degeneration is mediated via cytoplasmic L1-reverse-transcribed Alu cDNA independently of retrotransposition. Alu RNA did not induce cDNA production or RPE degeneration in L1-inhibited animals or human cells. Alu reverse transcription can be initiated in the cytoplasm via self-priming of Alu RNA. In four health insurance databases, use of nucleoside RT inhibitors was associated with reduced risk of developing atrophic macular degeneration (pooled adjusted hazard ratio, 0.616; 95% confidence interval, 0.493-0.770), thus identifying inhibitors of this Alu replication cycle shunt as potential therapies for a major cause of blindness.


Subject(s)
Alu Elements/genetics , Long Interspersed Nucleotide Elements/genetics , Macular Degeneration/genetics , Retinal Pigments/metabolism , Animals , Cytoplasm/genetics , DNA, Complementary/genetics , Epithelium/metabolism , Epithelium/pathology , Humans , Macular Degeneration/pathology , Retinal Pigments/biosynthesis , Retroelements/genetics , Reverse Transcription/genetics
6.
Invest Ophthalmol Vis Sci ; 61(10): 4, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32749462

ABSTRACT

Purpose: Azidothymidine (AZT), a nucleoside reverse transcriptase inhibitor, possesses anti-inflammatory and anti-angiogenic activity independent of its ability to inhibit reverse transcriptase. The aim of this study was to evaluate the efficacy of 5'-glucuronyl azidothymidine (GAZT), an antiretrovirally inert hepatic clinical metabolite of AZT, in mouse models of retinal pigment epithelium (RPE) degeneration and choroidal neovascularization (CNV), hallmark features of dry and wet age-related macular degeneration (AMD), respectively. Methods: RPE degeneration was induced in wild-type (WT) C57BL/6J mice by subretinal injection of Alu RNA. RPE degeneration was assessed by fundus photography and confocal microscopy of zonula occludens-1-stained RPE flat mounts. Choroidal neovascularization was induced by laser injury in WT mice, and CNV volume was measured by confocal microscopy. AZT and GAZT were delivered by intravitreous injections. Inflammasome activation was monitored by western blotting for caspase-1 and by ELISA for IL-1ß in Alu RNA-treated bone marrow-derived macrophages (BMDMs). Results: GAZT inhibited Alu RNA-induced RPE degeneration and laser-induced CNV. GAZT also reduced Alu RNA-induced caspase-1 activation and IL-1ß release in BMDMs. Conclusions: GAZT possesses dual anti-inflammatory and anti-angiogenic properties and could be a viable treatment option for both forms of AMD.


Subject(s)
Choroidal Neovascularization/drug therapy , Disease Models, Animal , Geographic Atrophy/drug therapy , Retinal Pigment Epithelium/drug effects , Reverse Transcriptase Inhibitors/therapeutic use , Zidovudine/analogs & derivatives , Animals , Blotting, Western , Caspase 1/metabolism , Choroidal Neovascularization/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Geographic Atrophy/metabolism , Interleukin-1beta/metabolism , Intravitreal Injections , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Reverse Transcriptase Inhibitors/administration & dosage , Zidovudine/administration & dosage , Zidovudine/therapeutic use , Zonula Occludens-1 Protein/metabolism
7.
Ophthalmology ; 126(12): 1667-1674, 2019 12.
Article in English | MEDLINE | ID: mdl-31281056

ABSTRACT

PURPOSE: To evaluate the relationship between OCT features and progression to late age related-macular degeneration (AMD) in the fellow eyes of patients enrolled in the Study of Ranibizumab Administered Monthly or on an As-needed Basis in Patients With Subfoveal Neovascular AMD (HARBOR) (ClinicalTrials.gov identifier, NCT00891735). DESIGN: Post hoc analysis of a phase 3 multicenter, prospective, randomized, double-masked, active treatment-controlled clinical trial. PARTICIPANTS: Evaluable patients (n = 501) with macular neovascularization (MNV) secondary to neovascular AMD and early or intermediate AMD in the fellow eye. METHODS: Volume OCT scans from 501 fellow eyes of 501 patients with MNV were reviewed. Baseline OCT features that were assessed included intraretinal hypereflective foci (IHRF), hyporeflective foci (hRF) within drusenoid lesions (DLs), subretinal drusenoid deposits (SDDs), and drusen volume (DV) of 0.03 mm3 or more. OCT images obtained at months 6, 12, 18, and 24 were graded by masked graders for late AMD (defined as MNV, complete retinal pigment epithelium and photoreceptor atrophy [cRORA], or both). Participant demographic characteristics (age, gender, and smoke exposure) and baseline OCT features were correlated with progression to late AMD. MAIN OUTCOME MEASURES: Incidence of late AMD, hazard ratio (HR) for demographics, and OCT risk factors. RESULTS: At month 24, 33.13% of eyes (166/501) demonstrated late AMD: 20.96% (105/501) demonstrated cRORA, whereas 12.18% (61/501) demonstrated MNV. Baseline demographic factors were not associated significantly with development of late AMD, whereas significant associations were identified for all OCT features. Intraretinal hypereflective foci had an HR of 5.21 (95% confidence interval [CI], 3.29-8.26), hRF within DLs had an HR of 2.42 (95% CI, 1.74-3.38), SDD had an HR of 1.95 (95% CI, 1.34-2.82), and DV of 0.03 mm3 or more had an HR of 1.46 (95% CI, 1.03-2.07). The correlation remained significant when considering only the progression to cRORA and MNV alone, except for DV, which was not associated significantly with progression to MNV. CONCLUSIONS: We confirmed that 4 previously reported OCT risk factors were associated with progression to late AMD in the fellow eyes of patients newly diagnosed with MNV. Although outcomes of more than 2 years were not evaluated, these findings may help to identify high-risk AMD patients.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Choroidal Neovascularization/diagnostic imaging , Ranibizumab/therapeutic use , Wet Macular Degeneration/diagnostic imaging , Aged , Aged, 80 and over , Choroidal Neovascularization/drug therapy , Disease Progression , Double-Blind Method , Female , Fluorescein Angiography , Humans , Incidence , Intravitreal Injections , Male , Proportional Hazards Models , Prospective Studies , Retinal Drusen/diagnostic imaging , Risk Factors , Tomography, Optical Coherence , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Visual Acuity , Wet Macular Degeneration/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...