Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Proc Natl Acad Sci U S A ; 117(5): 2579-2587, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31964819

ABSTRACT

Degeneration of the retinal pigmented epithelium (RPE) and aberrant blood vessel growth in the eye are advanced-stage processes in blinding diseases such as age-related macular degeneration (AMD), which affect hundreds of millions of people worldwide. Loss of the RNase DICER1, an essential factor in micro-RNA biogenesis, is implicated in RPE atrophy. However, the functional implications of DICER1 loss in choroidal and retinal neovascularization are unknown. Here, we report that two independent hypomorphic mouse strains, as well as a separate model of postnatal RPE-specific DICER1 ablation, all presented with spontaneous RPE degeneration and choroidal and retinal neovascularization. DICER1 hypomorphic mice lacking critical inflammasome components or the innate immune adaptor MyD88 developed less severe RPE atrophy and pathological neovascularization. DICER1 abundance was also reduced in retinas of the JR5558 mouse model of spontaneous choroidal neovascularization. Finally, adenoassociated vector-mediated gene delivery of a truncated DICER1 variant (OptiDicer) reduced spontaneous choroidal neovascularization in JR5558 mice. Collectively, these findings significantly expand the repertoire of DICER1 in preserving retinal homeostasis by preventing both RPE degeneration and pathological neovascularization.


Subject(s)
DEAD-box RNA Helicases/metabolism , Macular Degeneration/metabolism , Retinal Pigment Epithelium/blood supply , Ribonuclease III/metabolism , Animals , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Choroidal Neovascularization/physiopathology , DEAD-box RNA Helicases/genetics , Humans , Macular Degeneration/genetics , Macular Degeneration/pathology , Macular Degeneration/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/physiopathology , Retinal Neovascularization/genetics , Retinal Neovascularization/metabolism , Retinal Neovascularization/parasitology , Retinal Neovascularization/physiopathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Ribonuclease III/genetics
2.
Nat Med ; 24(1): 50-61, 2018 01.
Article in English | MEDLINE | ID: mdl-29176737

ABSTRACT

Geographic atrophy is a blinding form of age-related macular degeneration characterized by retinal pigmented epithelium (RPE) death; the RPE also exhibits DICER1 deficiency, resultant accumulation of endogenous Alu-retroelement RNA, and NLRP3-inflammasome activation. How the inflammasome is activated in this untreatable disease is largely unknown. Here we demonstrate that RPE degeneration in human-cell-culture and mouse models is driven by a noncanonical-inflammasome pathway that activates caspase-4 (caspase-11 in mice) and caspase-1, and requires cyclic GMP-AMP synthase (cGAS)-dependent interferon-ß production and gasdermin D-dependent interleukin-18 secretion. Decreased DICER1 levels or Alu-RNA accumulation triggers cytosolic escape of mitochondrial DNA, which engages cGAS. Moreover, caspase-4, gasdermin D, interferon-ß, and cGAS levels were elevated in the RPE in human eyes with geographic atrophy. Collectively, these data highlight an unexpected role of cGAS in responding to mobile-element transcripts, reveal cGAS-driven interferon signaling as a conduit for mitochondrial-damage-induced inflammasome activation, expand the immune-sensing repertoire of cGAS and caspase-4 to noninfectious human disease, and identify new potential targets for treatment of a major cause of blindness.


Subject(s)
Geographic Atrophy/enzymology , Inflammasomes/metabolism , Nucleotidyltransferases/metabolism , Animals , DEAD-box RNA Helicases/genetics , Humans , Interferon Type I/metabolism , Mice , Retinal Pigment Epithelium/metabolism , Ribonuclease III/genetics , Signal Transduction
3.
Article in English | MEDLINE | ID: mdl-26925256

ABSTRACT

Human intravenous immune globulin (IVIg), a purified IgG fraction composed of ~ 60% IgG1 and obtained from the pooled plasma of thousands of donors, is clinically used for a wide range of diseases. The biological actions of IVIg are incompletely understood and have been attributed both to the polyclonal antibodies therein and also to their IgG (IgG) Fc regions. Recently, we demonstrated that multiple therapeutic human IgG1 antibodies suppress angiogenesis in a target-independent manner via FcγRI, a high-affinity receptor for IgG1. Here we show that IVIg possesses similar anti-angiogenic activity and inhibited blood vessel growth in five different mouse models of prevalent human diseases, namely, neovascular age-related macular degeneration, corneal neovascularization, colorectal cancer, fibrosarcoma and peripheral arterial ischemic disease. Angioinhibition was mediated by the Fc region of IVIg, required FcγRI and had similar potency in transgenic mice expressing human FcγRs. Finally, IVIg therapy administered to humans for the treatment of inflammatory or autoimmune diseases reduced kidney and muscle blood vessel densities. These data place IVIg, an agent approved by the US Food and Drug Administration, as a novel angioinhibitory drug in doses that are currently administered in the clinical setting. In addition, they raise the possibility of an unintended effect of IVIg on blood vessels.

4.
Article in English | MEDLINE | ID: mdl-26918197

ABSTRACT

Aberrant angiogenesis is implicated in diseases affecting nearly 10% of the world's population. The most widely used anti-angiogenic drug is bevacizumab, a humanized IgG1 monoclonal antibody that targets human VEGFA. Although bevacizumab does not recognize mouse Vegfa, it inhibits angiogenesis in mice. Here we show bevacizumab suppressed angiogenesis in three mouse models not via Vegfa blockade but rather Fc-mediated signaling through FcγRI (CD64) and c-Cbl, impairing macrophage migration. Other approved humanized or human IgG1 antibodies without mouse targets (adalimumab, alemtuzumab, ofatumumab, omalizumab, palivizumab and tocilizumab), mouse IgG2a, and overexpression of human IgG1-Fc or mouse IgG2a-Fc, also inhibited angiogenesis in wild-type and FcγR humanized mice. This anti-angiogenic effect was abolished by Fcgr1 ablation or knockdown, Fc cleavage, IgG-Fc inhibition, disruption of Fc-FcγR interaction, or elimination of FcRγ-initated signaling. Furthermore, bevacizumab's Fc region potentiated its anti-angiogenic activity in humanized VEGFA mice. Finally, mice deficient in FcγRI exhibited increased developmental and pathological angiogenesis. These findings reveal an unexpected anti-angiogenic function for FcγRI and a potentially concerning off-target effect of hIgG1 therapies.

5.
Cell Rep ; 11(11): 1686-93, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26074074

ABSTRACT

Excess iron induces tissue damage and is implicated in age-related macular degeneration (AMD). Iron toxicity is widely attributed to hydroxyl radical formation through Fenton's reaction. We report that excess iron, but not other Fenton catalytic metals, induces activation of the NLRP3 inflammasome, a pathway also implicated in AMD. Additionally, iron-induced degeneration of the retinal pigmented epithelium (RPE) is suppressed in mice lacking inflammasome components caspase-1/11 or Nlrp3 or by inhibition of caspase-1. Iron overload increases abundance of RNAs transcribed from short interspersed nuclear elements (SINEs): Alu RNAs and the rodent equivalent B1 and B2 RNAs, which are inflammasome agonists. Targeting Alu or B2 RNA prevents iron-induced inflammasome activation and RPE degeneration. Iron-induced SINE RNA accumulation is due to suppression of DICER1 via sequestration of the co-factor poly(C)-binding protein 2 (PCBP2). These findings reveal an unexpected mechanism of iron toxicity, with implications for AMD and neurodegenerative diseases associated with excess iron.


Subject(s)
Alu Elements , Carrier Proteins/metabolism , Iron/toxicity , Retinal Pigment Epithelium/metabolism , Animals , Carrier Proteins/genetics , Caspase 1/genetics , Caspase 1/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Inflammasomes/metabolism , Iron/pharmacology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Retinal Pigment Epithelium/drug effects , Ribonuclease III/genetics , Ribonuclease III/metabolism
6.
Oncotarget ; 6(12): 10563-76, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25868854

ABSTRACT

To assess the therapeutic outcome of selective block of VEGFR1, we have evaluated the activity of a new specific antagonist of VEGFR1, named iVR1 (inhibitor of VEGFR1), in syngenic and xenograft colorectal cancer models, in an artificial model of metastatization, and in laser-induced choroid neovascularization. iVR1 inhibited tumor growth and neoangiogenesis in both models of colorectal cancer, with an extent similar to that of bevacizumab, a monoclonal antibody anti-VEGF-A. It potently inhibited VEGFR1 phosphorylation in vivo, determining a strong inhibition of the recruitment of monocyte-macrophages and of mural cells as confirmed, in vitro, by the ability to inhibit macrophages migration. iVR1 was able to synergize with irinotecan determining a shrinkage of tumors that became undetectable after three weeks of combined treatment. Such treatment induced a significant prolongation of survival similar to that observed with bevacizumab and irinotecan combination. iVR1 also fully prevented lung invasion by HCT-116 cells injected in mouse tail vein. Also, iVR1 impressively inhibited choroid neovascularization after a single intravitreal injection. Collectively, data showed the strong potential of iVR1 peptide as a new anti-tumor and anti-metastatic agent and demonstrate the high flexibility of VEGFR1 antagonists as therapeutic anti-angiogenic agents in different pathological contexts.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Colorectal Neoplasms/drug therapy , Oligopeptides/pharmacology , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/pathology , HCT116 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , RAW 264.7 Cells , Random Allocation , Xenograft Model Antitumor Assays
8.
Science ; 346(6212): 1000-3, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25414314

ABSTRACT

Nucleoside reverse transcriptase inhibitors (NRTIs) are mainstay therapeutics for HIV that block retrovirus replication. Alu (an endogenous retroelement that also requires reverse transcriptase for its life cycle)-derived RNAs activate P2X7 and the NLRP3 inflammasome to cause cell death of the retinal pigment epithelium in geographic atrophy, a type of age-related macular degeneration. We found that NRTIs inhibit P2X7-mediated NLRP3 inflammasome activation independent of reverse transcriptase inhibition. Multiple approved and clinically relevant NRTIs prevented caspase-1 activation, the effector of the NLRP3 inflammasome, induced by Alu RNA. NRTIs were efficacious in mouse models of geographic atrophy, choroidal neovascularization, graft-versus-host disease, and sterile liver inflammation. Our findings suggest that NRTIs are ripe for drug repurposing in P2X7-driven diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Inflammasomes/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Alu Elements , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Apoptosis/drug effects , Carrier Proteins/metabolism , Caspase 1/metabolism , Choroidal Neovascularization/drug therapy , Disease Models, Animal , Geographic Atrophy/drug therapy , Graft vs Host Disease/drug therapy , Hepatitis/drug therapy , Liver/drug effects , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Purinergic P2X7/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/physiology , Reverse Transcriptase Inhibitors/therapeutic use
9.
Proc Natl Acad Sci U S A ; 111(45): 16082-7, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25349431

ABSTRACT

Geographic atrophy, an advanced form of age-related macular degeneration (AMD) characterized by death of the retinal pigmented epithelium (RPE), causes untreatable blindness in millions worldwide. The RPE of human eyes with geographic atrophy accumulates toxic Alu RNA in response to a deficit in the enzyme DICER1, which in turn leads to activation of the NLRP3 inflammasome and elaboration of IL-18. Despite these recent insights, it is still unclear how RPE cells die during the course of the disease. In this study, we implicate the involvement of Caspase-8 as a critical mediator of RPE degeneration. Here we show that DICER1 deficiency, Alu RNA accumulation, and IL-18 up-regulation lead to RPE cell death via activation of Caspase-8 through a Fas ligand-dependent mechanism. Coupled with our observation of increased Caspase-8 expression in the RPE of human eyes with geographic atrophy, our findings provide a rationale for targeting this apoptotic pathway in this disease.


Subject(s)
Alu Elements , Apoptosis , Caspase 8/metabolism , DEAD-box RNA Helicases/metabolism , Eye Proteins/metabolism , Macular Degeneration/metabolism , RNA/metabolism , Ribonuclease III/metabolism , Animals , Caspase 8/genetics , DEAD-box RNA Helicases/genetics , Eye Proteins/genetics , Humans , Interleukin-18/genetics , Interleukin-18/metabolism , Macular Degeneration/pathology , Mice , Mice, Knockout , RNA/genetics , Ribonuclease III/genetics , Up-Regulation/genetics
10.
GMS Ophthalmol Cases ; 4: Doc02, 2014.
Article in English | MEDLINE | ID: mdl-27625937

ABSTRACT

PURPOSE: To report an atypically severe and refractory phlyctenular keratoconjunctivitis case treated successfully with systemic biologic immunosuppressive therapy. METHODS: A 10-year-old female was followed in the ophthalmology clinic for three years for a severe form of bilateral PKC. The patient was treated for blepharitis and intestinal parasitosis, and underwent topical corticosteroid therapy, followed by subconjunctival injections and systemic corticosteroids with no clinical improvement. An association of topical cyclosporine A and oral methotrexate had no clinical response either. Phlyctenae of the cornea remained evident with neovascularization, progressive peripheral corneal thinning and occasional anterior chamber reaction. RESULTS: The patient was treated with a combination of infliximab and methotrexate and corticosteroid therapy was tapered, with a fast and sustained resolution of the symptoms and corneal signs. Eleven months past initiation of the treatment, the patient remains asymptomatic and without any recurrence of the disease. CONCLUSION: Phlyctenular keratoconjunctivitis may present with a broad spectrum of symptoms and signs, and its severity varies significantly. In cases of severe PKC, which are refractory to conventional therapy, systemic biologic immunosuppressive therapy may be a valuable alternative.

11.
Invest Ophthalmol Vis Sci ; 54(12): 7395-401, 2013 Nov 11.
Article in English | MEDLINE | ID: mdl-24114535

ABSTRACT

PURPOSE: Accumulation of Alu RNA transcripts due to DICER1 deficiency in the retinal pigmented epithelium (RPE) promotes geographic atrophy. Recently we showed that Alu RNA activated the NLRP3 inflammasome, leading to RPE cell death via interleukin-18 (IL-18)-mediated MyD88 signaling. However, the molecular basis for NLRP3 inflammasome activation by Alu RNA is not well understood. We sought to decipher the key signaling events triggered by Alu RNA that lead to priming and activation of the NLRP3 inflammasome and, ultimately, to RPE degeneration by investigating the roles of the purinoreceptor P2X7, the transcription factor NF-κB, and the Toll-like receptors (TLRs) in these processes. METHODS: Human and mouse RPE cells were transfected with a plasmid encoding an Alu element (pAlu) or an in vitro-transcribed Alu RNA. Inflammasome priming was assessed by measuring NLRP3 and IL18 mRNA levels by real-time quantitative PCR. Using immunoblotting, we assessed NF-κB activation by monitoring phosphorylation of its p65 subunit, and inflammasome activation by monitoring caspase-1 cleavage into its active form. RPE degeneration was induced in mice by subretinal transfection of pAlu or Alu RNA. The NF-κB inhibitor BAY 11-7082, the P2X7 receptor antagonist A-740003, and the NLRP3 inflammasome inhibitor glyburide were delivered by intravitreous injections. We studied wild-type (WT) C57Bl/6J, P2rx7(-/-), Nfkb1(-/-), and Tlr23479(-/-) mice. RPE degeneration was assessed by fundus photography and zonula occludens-1 (ZO-1) staining of mouse RPE. RESULTS: Alu RNA-induced NF-κB activation, independent of TLR-1, -2, -3, -4, -6, -7, and -9 signaling, was required for priming the NLRP3 inflammasome. Nfkb1(-/-) and P2rx7(-/-) mice and WT mice treated with the pharmacological inhibitors of NF-κB, P2X7, or NLRP3, were protected against Alu RNA-induced RPE degeneration. CONCLUSIONS: NF-κB and P2X7 are critical signaling intermediates in Alu RNA-induced inflammasome priming and RPE degeneration. These molecules are novel targets for rational drug development for geographic atrophy.


Subject(s)
Alu Elements , Geographic Atrophy/physiopathology , Inflammasomes/physiology , Receptors, Purinergic P2X7/physiology , Signal Transduction/physiology , Toll-Like Receptors/physiology , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Caspase 1/metabolism , Disease Models, Animal , Glyburide/pharmacology , Humans , Interleukin-18/metabolism , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nitriles/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , RNA, Messenger/metabolism , Retinal Pigment Epithelium/cytology , Sulfones/pharmacology
12.
Elife ; 2: e00324, 2013 Jun 18.
Article in English | MEDLINE | ID: mdl-23795287

ABSTRACT

Optimal phototransduction requires separation of the avascular photoreceptor layer from the adjacent vascularized inner retina and choroid. Breakdown of peri-photoreceptor vascular demarcation leads to retinal angiomatous proliferation or choroidal neovascularization, two variants of vascular invasion of the photoreceptor layer in age-related macular degeneration (AMD), the leading cause of irreversible blindness in industrialized nations. Here we show that sFLT-1, an endogenous inhibitor of vascular endothelial growth factor A (VEGF-A), is synthesized by photoreceptors and retinal pigment epithelium (RPE), and is decreased in human AMD. Suppression of sFLT-1 by antibodies, adeno-associated virus-mediated RNA interference, or Cre/lox-mediated gene ablation either in the photoreceptor layer or RPE frees VEGF-A and abolishes photoreceptor avascularity. These findings help explain the vascular zoning of the retina, which is critical for vision, and advance two transgenic murine models of AMD with spontaneous vascular invasion early in life. DOI:http://dx.doi.org/10.7554/eLife.00324.001.


Subject(s)
Choroidal Neovascularization/metabolism , Macular Degeneration/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retinal Neovascularization/metabolism , Retinal Pigment Epithelium/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vision, Ocular , Adult , Aged , Aged, 80 and over , Animals , Antibodies/pharmacology , Case-Control Studies , Choroidal Neovascularization/genetics , Choroidal Neovascularization/pathology , Disease Models, Animal , Down-Regulation , Female , Humans , Macular Degeneration/genetics , Macular Degeneration/pathology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Photoreceptor Cells, Vertebrate/pathology , RNA Interference , Retinal Neovascularization/genetics , Retinal Neovascularization/pathology , Retinal Pigment Epithelium/pathology , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/deficiency , Vascular Endothelial Growth Factor Receptor-1/genetics
13.
PLoS One ; 7(12): e52450, 2012.
Article in English | MEDLINE | ID: mdl-23285048

ABSTRACT

Delta-like 4 (Dll4) is a ligand of the Notch pathway family which has been widely studied in the context of tumor angiogenesis, its blockade shown to result in non-productive angiogenesis and halted tumor growth. As Dll4 inhibitors enter the clinic, there is an emerging need to understand their side effects, namely the systemic consequences of Dll4:Notch blockade in tissues other than tumors. The present study focused on the effects of systemic anti-Dll4 targeting in the bone marrow (BM) microenvironment. Here we show that Dll4 blockade with monoclonal antibodies perturbs the BM vascular niche of sub-lethally irradiated mice, resulting in increased CD31(+), VE-Cadherin(+) and c-kit(+) vessel density, and also increased megakaryocytes, whereas CD105(+), VEGFR3(+), SMA(+) and lectin(+) vessel density remained unaltered. We investigated also the expression of angiocrine genes upon Dll4 treatment in vivo, and demonstrate that IGFbp2, IGFbp3, Angpt2, Dll4, DHH and VEGF-A are upregulated, while FGF1 and CSF2 are reduced. In vitro treatment of endothelial cells with anti-Dll4 reduced Akt phosphorylation while maintaining similar levels of Erk 1/2 phosphorylation. Besides its effects in the BM vascular niche, anti-Dll4 treatment perturbed hematopoiesis, as evidenced by increased myeloid (CD11b(+)), decreased B (B220(+)) and T (CD3(+)) lymphoid BM content of treated mice, with a corresponding increase in myeloid circulating cells. Moreover, anti-Dll4 treatment also increased the number of CFU-M and -G colonies in methylcellulose assays, independently of Notch1. Finally, anti-Dll4 treatment of donor BM improved the hematopoietic recovery of lethally irradiated recipients in a transplant setting. Together, our data reveals the hematopoietic (BM) effects of systemic anti-Dll4 treatment result from qualitative vascular changes and also direct hematopoietic cell modulation, which may be favorable in a transplant setting.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow/metabolism , Cellular Microenvironment , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Antibodies, Neutralizing/pharmacology , Bone Marrow/blood supply , Bone Marrow/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Transplantation , Calcium-Binding Proteins , Cellular Microenvironment/drug effects , Hematopoiesis/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Whole-Body Irradiation
SELECTION OF CITATIONS
SEARCH DETAIL
...