Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Invest Ophthalmol Vis Sci ; 64(5): 3, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37129905

RESUMEN

Purpose: Rhegmatogenous retinal detachment (RRD) is a vision-threatening event that benefits from surgical intervention. While awaiting surgical reattachment, irreversible hypoxic and inflammatory damage to the retina often occurs. An interim therapy protecting photoreceptors could improve functional outcomes. We sought to determine whether Kamuvudine-9 (K-9), a derivative of nucleoside reverse transcriptase inhibitors (NRTIs) that inhibits inflammasome activation, and the NRTIs lamivudine (3TC) and azidothymidine (AZT) could protect the retina following RRD. Methods: RRD was induced in mice via subretinal injection (SRI) of 1% carboxymethylcellulose (CMC). To simulate outcomes following the clinical management of RRD, we determined the optimal conditions by which SRI of CMC induced spontaneous retinal reattachment (SRR) occurs over 10 days (RRD/SRR). K-9, 3TC, or AZT was administered via intraperitoneal injection. Inflammasome activation pathways were monitored by abundance of cleaved caspase-1, IL-18, and cleaved caspase-8, and photoreceptor death was assessed by TUNEL staining. Retinal function was assessed by full-field scotopic electroretinography. Results: RRD induced retinal inflammasome activation and photoreceptor death in mice. Systemic administration of K-9, 3TC, or AZT inhibited retinal inflammasome activation and photoreceptor death. In the RRD/SRR model, K-9 protected retinal electrical function during the time of RRD and induced an improvement following retinal reattachment. Conclusions: K-9 and NRTIs exhibit anti-inflammatory and neuroprotective activities in experimental RRD. Given its capacity to protect photoreceptor function during the period of RRD and enhance retinal function following reattachment, K-9 shows promise as a retinal neuroprotectant and warrants study in RRD. Further, this novel RRD/SRR model may facilitate experimental evaluation of functional outcomes relevant to RRD.


Asunto(s)
Desprendimiento de Retina , Animales , Ratones , Desprendimiento de Retina/cirugía , Inflamasomas , Agudeza Visual , Retina , Estudios Retrospectivos , Vitrectomía
2.
medRxiv ; 2023 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-36993694

RESUMEN

Innate immune signaling through the NLRP3 inflammasome has been implicated in the pathogenesis of Alzheimer's disease (AD), the most prevalent form of dementia. We previously demonstrated that nucleoside reverse transcriptase inhibitors (NRTIs), drugs approved to treat HIV and hepatitis B infections, also inhibit inflammasome activation. Here we report that in humans, NRTI exposure was associated with a significantly lower incidence of AD in two of the largest health insurance databases in the United States. Treatment of aged 5xFAD mice (a mouse model of amyloid-ß deposition that expresses five mutations found in familial AD) with Kamuvudine-9 (K-9), an NRTI-derivative with enhanced safety profile, reduced Aß deposition and reversed their cognitive deficit by improving their spatial memory and learning performance to that of young wild-type mice. These findings support the concept that inflammasome inhibition could benefit AD and provide a rationale for prospective clinical testing of NRTIs or K-9 in AD.

3.
Sci Immunol ; 6(66): eabi4493, 2021 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-34860583

RESUMEN

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.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas de Unión al Calcio/inmunología , ARN Helicasas DEAD-box/inmunología , Inflamasomas/inmunología , ARN/inmunología , Retroelementos/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas de Unión al Calcio/deficiencia , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
Sci Adv ; 7(40): eabj3658, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34586848

RESUMEN

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.

5.
Signal Transduct Target Ther ; 6(1): 149, 2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33850097

RESUMEN

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.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Degeneración Macular/tratamiento farmacológico , Epitelio Pigmentado de la Retina/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología , Péptidos beta-Amiloides/genética , Animales , Modelos Animales de Enfermedad , Humanos , Degeneración Macular/genética , Degeneración Macular/metabolismo , Masculino , Ratones , Ratones Noqueados
6.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33526699

RESUMEN

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.


Asunto(s)
Elementos Alu/genética , Elementos de Nucleótido Esparcido Largo/genética , Degeneración Macular/genética , Pigmentos Retinianos/metabolismo , Animales , Citoplasma/genética , ADN Complementario/genética , Epitelio/metabolismo , Epitelio/patología , Humanos , Degeneración Macular/patología , Pigmentos Retinianos/biosíntesis , Retroelementos/genética , Transcripción Reversa/genética
7.
Nat Commun ; 11(1): 4737, 2020 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-32968070

RESUMEN

Innate immune signaling through the NLRP3 inflammasome is activated by multiple diabetes-related stressors, but whether targeting the inflammasome is beneficial for diabetes is still unclear. Nucleoside reverse-transcriptase inhibitors (NRTI), drugs approved to treat HIV-1 and hepatitis B infections, also block inflammasome activation. Here, we show, by analyzing five health insurance databases, that the adjusted risk of incident diabetes is 33% lower in patients with NRTI exposure among 128,861 patients with HIV-1 or hepatitis B (adjusted hazard ratio for NRTI exposure, 0.673; 95% confidence interval, 0.638 to 0.710; P < 0.0001; 95% prediction interval, 0.618 to 0.734). Meanwhile, an NRTI, lamivudine, improves insulin sensitivity and reduces inflammasome activation in diabetic and insulin resistance-induced human cells, as well as in mice fed with high-fat chow; mechanistically, inflammasome-activating short interspersed nuclear element (SINE) transcripts are elevated, whereas SINE-catabolizing DICER1 is reduced, in diabetic cells and mice. These data suggest the possibility of repurposing an approved class of drugs for prevention of diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Reposicionamiento de Medicamentos , Inflamasomas/efectos de los fármacos , Resistencia a la Insulina , Inhibidores de la Transcriptasa Inversa/farmacología , Adipocitos/metabolismo , Animales , Supervivencia Celular , ARN Helicasas DEAD-box/metabolismo , Diabetes Mellitus Tipo 2/prevención & control , Dieta Alta en Grasa/efectos adversos , VIH-1/efectos de los fármacos , Hepatitis B , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Células Musculares/metabolismo , Ribonucleasa III/metabolismo
8.
Invest Ophthalmol Vis Sci ; 61(10): 4, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32749462

RESUMEN

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.


Asunto(s)
Neovascularización Coroidal/tratamiento farmacológico , Modelos Animales de Enfermedad , Atrofia Geográfica/tratamiento farmacológico , Epitelio Pigmentado de la Retina/efectos de los fármacos , Inhibidores de la Transcriptasa Inversa/uso terapéutico , Zidovudina/análogos & derivados , Animales , Western Blotting , Caspasa 1/metabolismo , Neovascularización Coroidal/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Atrofia Geográfica/metabolismo , Interleucina-1beta/metabolismo , Inyecciones Intravítreas , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Inhibidores de la Transcriptasa Inversa/administración & dosificación , Zidovudina/administración & dosificación , Zidovudina/uso terapéutico , Proteína de la Zonula Occludens-1/metabolismo
10.
Nature ; 566(7742): 73-78, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30728521

RESUMEN

Retrotransposable elements are deleterious at many levels, and the failure of host surveillance systems for these elements can thus have negative consequences. However, the contribution of retrotransposon activity to ageing and age-associated diseases is not known. Here we show that during cellular senescence, L1 (also known as LINE-1) retrotransposable elements become transcriptionally derepressed and activate a type-I interferon (IFN-I) response. The IFN-I response is a phenotype of late senescence and contributes to the maintenance of the senescence-associated secretory phenotype. The IFN-I response is triggered by cytoplasmic L1 cDNA, and is antagonized by inhibitors of the L1 reverse transcriptase. Treatment of aged mice with the nucleoside reverse transcriptase inhibitor lamivudine downregulated IFN-I activation and age-associated inflammation (inflammaging) in several tissues. We propose that the activation of retrotransposons is an important component of sterile inflammation that is a hallmark of ageing, and that L1 reverse transcriptase is a relevant target for the treatment of age-associated disorders.


Asunto(s)
Senescencia Celular/genética , Inflamación/genética , Interferón Tipo I/metabolismo , Elementos de Nucleótido Esparcido Largo/genética , Envejecimiento/genética , Envejecimiento/patología , Animales , Regulación hacia Abajo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Inflamación/patología , Lamivudine/farmacología , Masculino , Ratones , Fenotipo , ADN Polimerasa Dirigida por ARN/genética , ADN Polimerasa Dirigida por ARN/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología
11.
Acta Neuropathol Commun ; 6(1): 27, 2018 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-29642926

RESUMEN

Duchenne muscular dystrophy (DMD) is the most common inherited muscle disorder that causes severe disability and death of young men. This disease is characterized by progressive muscle degeneration aggravated by sterile inflammation and is also associated with cognitive impairment and low bone density. Given that no current treatment can improve the long-term outcome, approaches with a strong translational potential are urgently needed. Duchenne muscular dystrophy (DMD) alters P2RX7 signaling in both muscle and inflammatory cells and inhibition of this receptor resulted in a significant attenuation of muscle and non-muscle symptoms in DMDmdx mouse model. As P2RX7 is an attractive target in a range of human diseases, specific antagonists have been developed. Yet, these will require lengthy safety testing in the pediatric population of Duchenne muscular dystrophy (DMD) patients. In contrast, Nucleoside Reverse Transcriptase Inhibitors (NRTIs) can act as P2RX7 antagonists and are drugs with an established safety record, including in children. We demonstrate here that AZT (Zidovudine) inhibits P2RX7 functions acting via the same allosteric site as other antagonists. Moreover, short-term AZT treatment at the peak of disease in DMDmdx mice attenuated the phenotype without any detectable side effects. Recovery was evident in the key parameters such as reduced sarcolemma permeability confirmed by lower serum creatine kinase levels and IgG influx into myofibres, decreased inflammatory cell numbers and inflammation markers in leg and heart muscles of treated mice. Moreover, this short-term therapy had some positive impact on muscle strength in vivo and no detrimental effect on mitochondria, which is the main side-effect of Nucleoside Reverse Transcriptase Inhibitors (NRTIs). Given these results, we postulate that AZT could be quickly re-purposed for the treatment of this highly debilitating and lethal disease. This approach is not constrained by causative DMD mutations and may be effective in alleviating both muscle and non-muscle abnormalities.


Asunto(s)
Antimetabolitos/uso terapéutico , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/terapia , Receptores Purinérgicos P2X7/metabolismo , Zidovudina/uso terapéutico , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Calcio/metabolismo , Células Cultivadas , Colágeno Tipo IV/metabolismo , Creatina Quinasa/sangre , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Modelos Moleculares , Fuerza Muscular/efectos de los fármacos , Músculos/efectos de los fármacos , Músculos/metabolismo , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/genética , Mioblastos/efectos de los fármacos
12.
Nat Med ; 24(1): 50-61, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29176737

RESUMEN

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.


Asunto(s)
Atrofia Geográfica/enzimología , Inflamasomas/metabolismo , Nucleotidiltransferasas/metabolismo , Animales , ARN Helicasas DEAD-box/genética , Humanos , Interferón Tipo I/metabolismo , Ratones , Epitelio Pigmentado de la Retina/metabolismo , Ribonucleasa III/genética , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...