Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 141(3): 271-284, 2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36351237

RESUMEN

Homeostatic adaptation to systemic iron overload involves transcriptional induction of bone morphogenetic protein 6 (BMP6) in liver sinusoidal endothelial cells (LSECs). BMP6 is then secreted to activate signaling of the iron hormone hepcidin (HAMP) in neighboring hepatocytes. To explore the mechanism of iron sensing by LSECs, we generated TfrcTek-Cre mice with endothelial cell-specific ablation of transferrin receptor 1 (Tfr1). We also used control Tfrcfl/fl mice to characterize the LSEC-specific molecular responses to iron using single-cell transcriptomics. TfrcTek-Cre animals tended to have modestly increased liver iron content (LIC) compared with Tfrcfl/fl controls but expressed physiological Bmp6 and Hamp messenger RNA (mRNA). Despite a transient inability to upregulate Bmp6, they eventually respond to iron challenges with Bmp6 and Hamp induction, yet occasionally to levels slightly lower relative to LIC. High dietary iron intake triggered the accumulation of serum nontransferrin bound iron (NTBI), which significantly correlated with liver Bmp6 and Hamp mRNA levels and elicited more profound alterations in the LSEC transcriptome than holo-transferrin injection. This culminated in the robust induction of Bmp6 and other nuclear factor erythroid 2-related factor 2 (Nrf2) target genes, as well as Myc target genes involved in ribosomal biogenesis and protein synthesis. LSECs and midzonal hepatocytes were the most responsive liver cells to iron challenges and exhibited the highest expression of Bmp6 and Hamp mRNAs, respectively. Our data suggest that during systemic iron overload, LSECs internalize NTBI, which promotes oxidative stress and thereby transcriptionally induces Bmp6 via Nrf2. Tfr1 appears to contribute to iron sensing by LSECs, mostly under low iron conditions.


Asunto(s)
Sobrecarga de Hierro , Hierro , Ratones , Animales , Hierro/metabolismo , Transferrina/metabolismo , Células Endoteliales/metabolismo , Proteína Morfogenética Ósea 6/genética , Proteína Morfogenética Ósea 6/metabolismo , Factor 2 Relacionado con NF-E2 , Hepatocitos/metabolismo , Hígado/metabolismo , Hepcidinas/genética , Hepcidinas/metabolismo , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/metabolismo , ARN Mensajero/metabolismo
2.
Artículo en Inglés | MEDLINE | ID: mdl-37805023

RESUMEN

BACKGROUND: Nicotinamide adenine dinucleotide phosphate oxidase complex 2 (NOX2) deficiency, or chronic granulomatous disease (CGD), is an inborn error of immunity associated with increased susceptibility to infection and inflammatory manifestations. The pathophysiologic mechanism leading to the increased inflammatory response in CGD remains elusive. OBJECTIVE: We investigated the pathophysiologic mechanisms leading to NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome activation in NOX2 deficiency. METHODS: We used NOX2-deficient human primary and CRISPR-engineered macrophages to show that NOX2 deficiency enhances the inflammatory response mainly by modulating the 2 steps of NLRP3 inflammasome activation: its transcriptional priming and its posttranslational triggering. RESULTS: At the transcriptional level, NOX2-deficient phagocytes display increased priming of the NLRP3 inflammasome, as evidenced by increased transcription of NLRP3 and IL-1ß through an IL-1ß-dependent stimulation of the nuclear factor kappa-light-chain enhancer of activated B cells (aka NF-κB) pathway. At the posttranslational level, the absence of NOX2 triggers the NLRP3 inflammasome activation by increased K+ efflux and excessive release of mitochondrial DNA due to mitochondrial damage. Furthermore, NLRP3-driven pyroptosis in NOX2-deficient phagocytes further enhances NLRP3 activation by increasing K+ efflux. CONCLUSION: Our results unveil the role of NOX2 as a repressor of the inflammatory response at both transcriptional and posttranslational levels and pave the way for a more targeted approach to treating CGD patients with inflammatory manifestations.

3.
Cell Commun Signal ; 21(1): 196, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37940970

RESUMEN

The GPCR HCAR1 is known to be the sole receptor for lactate, which modulates its metabolic effects. Despite its significant role in many processes, mice deficient in HCAR1 exhibit no visible phenotype and are healthy and fertile. We performed transcriptomic analysis on HCAR1 deficient cells, in combination with lactate, to explore pathophysiologically altered processes. Processes such as immune regulation, various cancers, and neurodegenerative diseases were significantly enriched for HCAR1 transcriptomic signature. However, the most affected process of all was autism spectrum disorder. We performed behavioral tests on HCAR1 KO mice and observed that these mice manifest autistic-like behavior. Our data opens new avenues for research on HCAR1 and lactate effect at a pathological level. Video Abstract.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Ratones , Animales , Ácido Láctico/metabolismo , Transducción de Señal , Receptores Acoplados a Proteínas G/metabolismo
4.
Cell Mol Life Sci ; 79(1): 37, 2021 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-34971428

RESUMEN

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.


Asunto(s)
Neovascularización Patológica , Neovascularización Fisiológica , Óxido Nítrico Sintasa de Tipo III/fisiología , Retina/metabolismo , Neovascularización Retiniana , Vasos Retinianos , Animales , Bovinos , Línea Celular , Movimiento Celular , Polaridad Celular , Células Endoteliales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Retina/citología , Retina/patología , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Vasos Retinianos/citología , Vasos Retinianos/patología
5.
Pediatr Crit Care Med ; 23(1): 22-33, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34593741

RESUMEN

OBJECTIVES: Our understanding of pediatric acute respiratory distress syndrome is based on information from studies reporting intermittent, serial respiratory data. We have analyzed a high-resolution, longitudinal dataset that incorporates measures of hypoxemia severity, metrics of lung mechanics, ventilatory ratio, and mechanical power and examined associations with survival after the onset of pediatric acute respiratory distress syndrome. DESIGN: Single-center retrospective cohort, 2013-2018. SETTING: Tertiary surgical/medical PICU. PATIENTS: Seventy-six cases of severe pediatric acute respiratory distress syndrome, determined according to the Pediatric Acute Lung Injury Consensus Conference criteria. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: The high-resolution database included continuous monitoring of ventilatory data (0.03 Hz) for up to 14 days after the diagnosis of pediatric acute respiratory distress syndrome or until extubation or death (n = 26). In the 12,128 hours of data during conventional mechanical ventilation, we used generalized estimating equations to compare groups, accounting for any effect of time. We identified an association between survival and faster rate of improvement in delta pressure (peak inspiratory pressure minus positive end-expiratory pressure; p = 0.028). Nonsurvival was associated with higher daily Pediatric Logistic Organ Dysfunction-2 scores (p = 0.005) and more severe hypoxemia metrics (p = 0.005). Mortality was also associated with the following respiratory/pulmonary metrics (mean difference [95% CI]): positive end-expiratory pressure level (+2.0 cm H2O [0.8-3.2 cm H2O]; p = 0.001), peak inspiratory pressure level (+3.0 cm H2O [0.5-5.5 cm H2O]; p = 0.022), respiratory rate (z scores +2.2 [0.9-3.6]; p = 0.003], ventilatory ratio (+0.41 [0.28-0.55]; p = 0.0001], and mechanical power (+5 Joules/min [1-10 Joules/min]; p = 0.013). Based on generalized linear mixed modeling, mechanical power remained associated with mortality after adjustment for normal respiratory rate, age, and daily Pediatric Logistic Organ Dysfunction-2 score (+3 Joules/breath [1-6 Joules/breath]; p = 0.009). CONCLUSIONS: Mortality after severe pediatric acute respiratory distress syndrome is associated with the severity of organ dysfunction, oxygenation defects, and pulmonary metrics including dead space and theoretical mechanical energy load.


Asunto(s)
Síndrome de Dificultad Respiratoria , Niño , Humanos , Pulmón , Respiración Artificial , Síndrome de Dificultad Respiratoria/terapia , Estudios Retrospectivos , Análisis de Secuencia
6.
Proc Natl Acad Sci U S A ; 116(10): 4538-4547, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30787185

RESUMEN

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.


Asunto(s)
Permeabilidad Capilar , Retinopatía Diabética/patología , Receptor Notch1/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Proteínas de Unión al Calcio/biosíntesis , Activación Enzimática , Hiperglucemia/metabolismo , Proteína Jagged-1/biosíntesis , Ratones , Óxido Nítrico/biosíntesis , Vasos Retinianos/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Familia-src Quinasas/metabolismo
7.
Diabetologia ; 64(1): 70-82, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33099660

RESUMEN

AIMS/HYPOTHESIS: Proliferative diabetic retinopathy (PDR) with retinal neovascularisation (NV) is a leading cause of vision loss. This study identified a set of metabolites that were altered in the vitreous humour of PDR patients compared with non-diabetic control participants. We corroborated changes in vitreous metabolites identified in prior studies and identified novel dysregulated metabolites that may lead to treatment strategies for PDR. METHODS: We analysed metabolites in vitreous samples from 43 PDR patients and 21 non-diabetic epiretinal membrane control patients from Japan (age 27-80 years) via ultra-high-performance liquid chromatography-mass spectrometry. We then investigated the association of a novel metabolite (creatine) with retinal NV in mouse oxygen-induced retinopathy (OIR). Creatine or vehicle was administered from postnatal day (P)12 to P16 (during induced NV) via oral gavage. P17 retinas were quantified for NV and vaso-obliteration. RESULTS: We identified 158 metabolites in vitreous samples that were altered in PDR patients vs control participants. We corroborated increases in pyruvate, lactate, proline and allantoin in PDR, which were identified in prior studies. We also found changes in metabolites not previously identified, including creatine. In human vitreous humour, creatine levels were decreased in PDR patients compared with epiretinal membrane control participants (false-discovery rate <0.001). We validated that lower creatine levels were associated with vascular proliferation in mouse retina in the OIR model (p = 0.027) using retinal metabolomics. Oral creatine supplementation reduced NV compared with vehicle (P12 to P16) in OIR (p = 0.0024). CONCLUSIONS/INTERPRETATION: These results suggest that metabolites from vitreous humour may reflect changes in metabolism that can be used to find pathways influencing retinopathy. Creatine supplementation could be useful to suppress NV in PDR. Graphical abstract.


Asunto(s)
Retinopatía Diabética/metabolismo , Metabolómica , Cuerpo Vítreo/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Aminoácidos/análisis , Animales , Cromatografía Líquida de Alta Presión , Creatina/administración & dosificación , Creatina/análisis , Retinopatía Diabética/fisiopatología , Femenino , Humanos , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neovascularización Retiniana/metabolismo , Cuerpo Vítreo/química
8.
J Neuroinflammation ; 17(1): 359, 2020 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-33246504

RESUMEN

BACKGROUND: Inflammation and particularly interleukin-1ß (IL-1ß), a pro-inflammatory cytokine highly secreted by activated immune cells during early AMD pathological events, contribute significantly to retinal neurodegeneration. Here, we identify specific cell types that generate IL-1ß and harbor the IL-1 receptor (IL-1R) and pharmacologically validate IL-1ß's contribution to neuro-retinal degeneration using the IL-1R allosteric modulator composed of the amino acid sequence rytvela (as well as the orthosteric antagonist, Kineret) in a model of blue light-induced retinal degeneration. METHODS: Mice were exposed to blue light for 6 h and sacrificed 3 days later. Mice were intraperitoneally injected with rytvela, Kineret, or vehicle twice daily for 3 days. The inflammatory markers F4/80, NLRP3, caspase-1, and IL-1ß were assessed in the retinas. Single-cell RNA sequencing was used to determine the cell-specific expression patterns of retinal Il1b and Il1r1. Macrophage-induced photoreceptor death was assessed ex vivo using retinal explants co-cultured with LPS-activated bone marrow-derived macrophages. Photoreceptor cell death was evaluated by the TUNEL assay. Retinal function was assessed by flash electroretinography. RESULTS: Blue light markedly increased the mononuclear phagocyte recruitment and levels of inflammatory markers associated with photoreceptor death. Co-localization of NLRP3, caspase-1, and IL-1ß with F4/80+ mononuclear phagocytes was clearly detected in the subretinal space, suggesting that these inflammatory cells are the main source of IL-1ß. Single-cell RNA sequencing confirmed the immune-specific expression of Il1b and notably perivascular macrophages in light-challenged mice, while Il1r1 expression was found primarily in astrocytes, bipolar, and vascular cells. Retinal explants co-cultured with LPS/ATP-activated bone marrow-derived macrophages displayed a high number of TUNEL-positive photoreceptors, which was abrogated by rytvela treatment. IL-1R antagonism significantly mitigated the inflammatory response triggered in vivo by blue light exposure, and rytvela was superior to Kineret in preserving photoreceptor density and retinal function. CONCLUSION: These findings substantiate the importance of IL-1ß in neuro-retinal degeneration and revealed specific sources of Il1b from perivascular MPs, with its receptor Ilr1 being separately expressed on surrounding neuro-vascular and astroglial cells. They also validate the efficacy of rytvela-induced IL-1R modulation in suppressing detrimental inflammatory responses and preserving photoreceptor density and function in these conditions, reinforcing the rationale for clinical translation.


Asunto(s)
Interleucina-1beta/inmunología , Péptidos/farmacología , Células Fotorreceptoras/patología , Receptores de Interleucina-1/antagonistas & inhibidores , Degeneración Retiniana/patología , Animales , Modelos Animales de Enfermedad , Inflamación/inmunología , Inflamación/patología , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Células Fotorreceptoras/efectos de los fármacos , Degeneración Retiniana/inmunología
9.
Am J Pathol ; 189(9): 1878-1896, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31220454

RESUMEN

Ischemic retinopathies are characterized by a progressive microvascular degeneration followed by a postischemic aberrant neovascularization. To reinstate vascular supply and metabolic equilibrium to the ischemic tissue during ischemic retinopathies, a dysregulated production of growth factors and metabolic intermediates occurs, promoting retinal angiogenesis. Glycolysis-derived lactate, highly produced during ischemic conditions, has been associated with tumor angiogenesis and wound healing. Lactate exerts its biological effects via G-protein-coupled receptor 81 (GPR81) in several tissues; however, its physiological functions and mechanisms of action in the retina remain poorly understood. Herein, we show that GPR81, localized predominantly in Müller cells, governs deep vascular complex formation during development and in ischemic retinopathy. Lactate-stimulated GPR81 Müller cells produce numerous angiogenic factors, including Wnt ligands and particularly Norrin, which contributes significantly in triggering inner retinal blood vessel formation. Conversely, GPR81-null mice retina shows reduced inner vascular network formation associated with low levels of Norrin (and Wnt ligands). Lactate accumulation during ischemic retinopathy selectively activates GPR81-extracellular signal-regulated kinase 1/2-Norrin signaling to accelerate inner retinal vascularization in wild-type animals, but not in the retina of GPR81-null mice. Altogether, we reveal that lactate via GPR81-Norrin participates in inner vascular network development and in restoration of the vasculature in response to injury. These findings suggest a new potential therapeutic target to alleviate ischemic diseases.


Asunto(s)
Células Ependimogliales/patología , Proteínas del Ojo/metabolismo , Isquemia/patología , Proteínas del Tejido Nervioso/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Enfermedades de la Retina/patología , Neovascularización Retiniana/patología , Vasos Retinianos/patología , Proteínas Wnt/metabolismo , Animales , Células Ependimogliales/metabolismo , Proteínas del Ojo/genética , Isquemia/etiología , Isquemia/metabolismo , Ácido Láctico/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Enfermedades de la Retina/etiología , Enfermedades de la Retina/metabolismo , Neovascularización Retiniana/etiología , Neovascularización Retiniana/metabolismo , Vasos Retinianos/metabolismo , Proteínas Wnt/genética
10.
J Immunol ; 198(5): 2047-2062, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28148737

RESUMEN

Preterm birth (PTB) is commonly accompanied by in utero fetal inflammation, and existing tocolytic drugs do not target fetal inflammatory injury. Of the candidate proinflammatory mediators, IL-1 appears central and is sufficient to trigger fetal loss. Therefore, we elucidated the effects of antenatal IL-1 exposure on postnatal development and investigated two IL-1 receptor antagonists, the competitive inhibitor anakinra (Kineret) and a potent noncompetitive inhibitor 101.10, for efficacy in blocking IL-1 actions. Antenatal exposure to IL-1ß induced Tnfa, Il6, Ccl2, Pghs2, and Mpges1 expression in placenta and fetal membranes, and it elevated amniotic fluid IL-1ß, IL-6, IL-8, and PGF2α, resulting in PTB and marked neonatal mortality. Surviving neonates had increased Il1b, Il6, Il8, Il10, Pghs2, Tnfa, and Crp expression in WBCs, elevated plasma levels of IL-1ß, IL-6, and IL-8, increased IL-1ß, IL-6, and IL-8 in fetal lung, intestine, and brain, and morphological abnormalities: e.g., disrupted lung alveolarization, atrophy of intestinal villus and colon-resident lymphoid follicle, and degeneration and atrophy of brain microvasculature with visual evoked potential anomalies. Late gestation treatment with 101.10 abolished these adverse outcomes, whereas Kineret exerted only modest effects and no benefit for gestation length, neonatal mortality, or placental inflammation. In a LPS-induced model of infection-associated PTB, 101.10 prevented PTB, neonatal mortality, and fetal brain inflammation. There was no substantive deviation in postnatal growth trajectory or adult body morphometry after antenatal 101.10 treatment. The results implicate IL-1 as an important driver of neonatal morbidity in PTB and identify 101.10 as a safe and effective candidate therapeutic.


Asunto(s)
Encéfalo/inmunología , Desarrollo Fetal/efectos de los fármacos , Inflamación/inmunología , Interleucina-1beta/inmunología , Placenta/inmunología , Embarazo/inmunología , Nacimiento Prematuro/inmunología , Animales , Animales Recién Nacidos , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación/tratamiento farmacológico , Mediadores de Inflamación/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Interleucina-1beta/antagonistas & inhibidores , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Péptidos/uso terapéutico , Placenta/efectos de los fármacos , Nacimiento Prematuro/tratamiento farmacológico
11.
Am J Pathol ; 185(2): 581-95, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25478809

RESUMEN

Ischemic retinopathies are characterized by sequential vaso-obliteration followed by abnormal intravitreal neovascularization predisposing patients to retinal detachment and blindness. Ischemic retinopathies are associated with robust inflammation that leads to generation of IL-1ß, which causes vascular degeneration and impairs retinal revascularization in part through the liberation of repulsive guidance cue semaphorin 3A (Sema3A). However, retinal revascularization begins as inflammation culminates in ischemic retinopathies. Because inflammation leads to activation of proteases involved in the formation of vasculature, we hypothesized that proteinase-activated receptor (Par)-2 (official name F2rl1) may modulate deleterious effects of IL-1ß. Par2, detected mostly in retinal ganglion cells, was up-regulated in oxygen-induced retinopathy. Surprisingly, oxygen-induced retinopathy-induced vaso-obliteration and neovascularization were unaltered in Par2 knockout mice, suggesting compensatory mechanisms. We therefore conditionally knocked down retinal Par2 with shRNA-Par2-encoded lentivirus. Par2 knockdown interfered with normal revascularization, resulting in pronounced intravitreal neovascularization; conversely, the Par2 agonist peptide (SLIGRL) accelerated normal revascularization. In vitro and in vivo exploration of mechanisms revealed that IL-1ß induced Par2 expression, which in turn down-regulated sequentially IL-1 receptor type I and Sema3A expression through Erk/Jnk-dependent processes. Collectively, our findings unveil an important mechanism by which IL-1ß regulates its own endothelial cytotoxic actions by augmenting neuronal Par2 expression to repress sequentially IL-1 receptor type I and Sema3A expression. Timely activation of Par2 may be a promising therapeutic avenue in ischemic retinopathies.


Asunto(s)
Proteínas del Ojo/metabolismo , Isquemia/metabolismo , Receptores de Trombina/metabolismo , Enfermedades de la Retina/metabolismo , Neuronas Retinianas/metabolismo , Animales , Proteínas del Ojo/agonistas , Proteínas del Ojo/genética , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Isquemia/tratamiento farmacológico , Isquemia/genética , Isquemia/patología , Ratones , Ratones Noqueados , Oligopéptidos/farmacología , Receptores de Trombina/agonistas , Receptores de Trombina/genética , Enfermedades de la Retina/tratamiento farmacológico , Enfermedades de la Retina/genética , Enfermedades de la Retina/patología , Neuronas Retinianas/patología , Semaforina-3A/genética , Semaforina-3A/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 34(3): 581-6, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24458713

RESUMEN

OBJECTIVE: Regulation of angiogenesis is critical for many diseases. Specifically, pathological retinal neovascularization, a major cause of blindness, is suppressed with dietary ω3-long-chain polyunsaturated fatty acids (ω3LCPUFAs) through antiangiogenic metabolites of cyclooxygenase and lipoxygenase. Cytochrome P450 epoxygenases (CYP2C8) also metabolize LCPUFAs, producing bioactive epoxides, which are inactivated by soluble epoxide hydrolase (sEH) to transdihydrodiols. The effect of these enzymes and their metabolites on neovascularization is unknown. APPROACH AND RESULTS: The mouse model of oxygen-induced retinopathy was used to investigate retinal neovascularization. We found that CYP2C (localized in wild-type monocytes/macrophages) is upregulated in oxygen-induced retinopathy, whereas sEH is suppressed, resulting in an increased retinal epoxide:diol ratio. With a ω3LCPUFA-enriched diet, retinal neovascularization increases in Tie2-driven human-CYP2C8-overexpressing mice (Tie2-CYP2C8-Tg), associated with increased plasma 19,20-epoxydocosapentaenoic acid and retinal epoxide:diol ratio. 19,20-Epoxydocosapentaenoic acids and the epoxide:diol ratio are decreased with overexpression of sEH (Tie2-sEH-Tg). Overexpression of CYP2C8 or sEH in mice does not change normal retinal vascular development compared with their wild-type littermate controls. The proangiogenic role in retina of CYP2C8 with both ω3LCPUFA and ω6LCPUFA and antiangiogenic role of sEH in ω3LCPUFA metabolism were corroborated in aortic ring assays. CONCLUSIONS: Our results suggest that CYP2C ω3LCPUFA metabolites promote retinal pathological angiogenesis. CYP2C8 is part of a novel lipid metabolic pathway influencing retinal neovascularization.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Ácidos Grasos Omega-3/toxicidad , Macrófagos/enzimología , Monocitos/enzimología , Neovascularización Retiniana/inducido químicamente , Animales , Ácido Araquidónico/metabolismo , Hidrocarburo de Aril Hidroxilasas/genética , Biotransformación , Hipoxia de la Célula , Citocromo P-450 CYP2C8 , Grasas de la Dieta/farmacocinética , Ácidos Docosahexaenoicos/metabolismo , Ácido Eicosapentaenoico/metabolismo , Epóxido Hidrolasas/deficiencia , Epóxido Hidrolasas/genética , Epóxido Hidrolasas/fisiología , Proteínas del Ojo/metabolismo , Ácidos Grasos Omega-3/administración & dosificación , Ácidos Grasos Omega-3/clasificación , Ácidos Grasos Omega-3/farmacocinética , Ácidos Grasos Insaturados/administración & dosificación , Ácidos Grasos Insaturados/farmacocinética , Humanos , Lipooxigenasa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxígeno/toxicidad , Prostaglandina-Endoperóxido Sintasas/metabolismo , ARN Mensajero/biosíntesis , Receptor TIE-2/genética , Proteínas Recombinantes de Fusión/metabolismo , Neovascularización Retiniana/prevención & control
13.
Arterioscler Thromb Vasc Biol ; 33(8): 1881-91, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23766263

RESUMEN

OBJECTIVE: Proinflammatory cytokines contribute to the development of retinal vasculopathies. However, the role of these factors and the mechanisms by which they elicit their effects in retina are not known. We investigated whether activated microglia during early stages of ischemic retinopathy produces excessive interleukin-1ß (IL-1ß), which elicits retinal microvascular degeneration not directly but rather by triggering the release of the proapoptotic/repulsive factor semaphorin-3A (Sema3A) from neurons. APPROACH AND RESULTS: Sprague Dawley rats subjected to retinopathy induced by hyperoxia (80% O2; O2-induced retinopathy) exhibited retinal vaso-obliteration associated with microglial activation, NLRP3 upregulation, and IL-1ß and Sema3A release; IL-1ß was mostly generated by microglia. Intraperitoneal administration of IL-1 receptor antagonists (Kineret, or rytvela [101.10]) decreased these effects and enhanced retinal revascularization; knockdown of Sema3A resulted in microvessel preservation and, conversely, administration of IL-1ß caused vaso-obliteration. In vitro, IL-1ß derived from activated primary microglial cells, cultured under hyperoxia, stimulated the release of Sema3A in retinal ganglion cells-5, which in turn induced apoptosis of microvascular endothelium; antagonism of IL-1 receptor decreased microglial activation and on retinal ganglion cells-5 abolished the release of Sema3A inhibiting ensuing endothelial cell apoptosis. IL-1ß was not directly cytotoxic to endothelial cells. CONCLUSIONS: Our findings suggest that in the early stages of O2-induced retinopathy, retinal microglia are activated to produce IL-1ß, which sustains the activation of microglia and induces microvascular injury through the release of Sema3A from adjacent neurons. Interference with IL-1 receptor or Sema3A actions preserves the microvascular bed in ischemic retinopathies and, consequently, decreases ensued pathological preretinal neovascularization.


Asunto(s)
Interleucina-1beta/metabolismo , Isquemia/patología , Microglía/patología , Enfermedades de la Retina/patología , Retinitis/patología , Semaforina-3A/metabolismo , Animales , Antirreumáticos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Proteínas Portadoras , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Técnicas de Silenciamiento del Gen , Hiperoxia/inmunología , Hiperoxia/metabolismo , Hiperoxia/patología , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/inmunología , Isquemia/tratamiento farmacológico , Isquemia/inmunología , Microcirculación/fisiología , Microglía/inmunología , Microglía/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Péptidos/farmacología , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/inmunología , Receptores Citoplasmáticos y Nucleares/metabolismo , Enfermedades de la Retina/tratamiento farmacológico , Enfermedades de la Retina/inmunología , Células Ganglionares de la Retina/inmunología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Retinitis/tratamiento farmacológico , Retinitis/inmunología , Semaforina-3A/genética , Semaforina-3A/inmunología
15.
Mol Genet Metab Rep ; 39: 101073, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38550975

RESUMEN

An 11-month-old girl with severe acidosis, lethargy and vomiting, was diagnosed with holocarboxylase synthetase deficiency. She received biotin and was stable until age 8 years when vomiting, severe acidosis, hypoglycemia, and hyperammonemia developed. Management with intravenous glucose aiming to stimulate anabolism led to hyperglycemic ketoacidosis. Insulin therapy rapidly corrected biochemical parameters, and clinical status improved. We propose that secondary Krebs cycle disturbances affecting pancreatic beta cells impaired glucose-stimulated insulin secretion, resulting in insulinopenia.

16.
Cell Rep ; 43(1): 113616, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38150367

RESUMEN

Macrophages populate the embryo early in gestation, but their role in development is not well defined. In particular, specification and function of macrophages in intestinal development remain little explored. To study this event in the human developmental context, we derived and combined human intestinal organoid and macrophages from pluripotent stem cells. Macrophages migrate into the organoid, proliferate, and occupy the emerging microanatomical niches of epithelial crypts and ganglia. They also acquire a transcriptomic profile similar to that of fetal intestinal macrophages and display tissue macrophage behaviors, such as recruitment to tissue injury. Using this model, we show that macrophages reduce glycolysis in mesenchymal cells and limit tissue growth without affecting tissue architecture, in contrast to the pro-growth effect of enteric neurons. In short, we engineered an intestinal tissue model populated with macrophages, and we suggest that resident macrophages contribute to the regulation of metabolism and growth of the developing intestine.


Asunto(s)
Macrófagos , Células Madre Pluripotentes , Humanos , Diferenciación Celular , Macrófagos/metabolismo , Intestinos , Células Madre Pluripotentes/metabolismo , Intestino Delgado , Organoides/metabolismo
17.
Nat Commun ; 15(1): 4405, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38782923

RESUMEN

Zonula occludens-1 (ZO-1) is involved in the regulation of cell-cell junctions between endothelial cells (ECs). Here we identify the ZO-1 protein interactome and uncover ZO-1 interactions with RNA-binding proteins that are part of stress granules (SGs). Downregulation of ZO-1 increased SG formation in response to stress and protected ECs from cellular insults. The ZO-1 interactome uncovered an association between ZO-1 and Y-box binding protein 1 (YB-1), a constituent of SGs. Arsenite treatment of ECs decreased the interaction between ZO-1 and YB-1, and drove SG assembly. YB-1 expression is essential for SG formation and for the cytoprotective effects induced by ZO-1 downregulation. In the developing retinal vascular plexus of newborn mice, ECs at the front of growing vessels express less ZO-1 but display more YB-1-positive granules than ECs located in the vascular plexus. Endothelial-specific deletion of ZO-1 in mice at post-natal day 7 markedly increased the presence of YB-1-positive granules in ECs of retinal blood vessels, altered tip EC morphology and vascular patterning, resulting in aberrant endothelial proliferation, and arrest in the expansion of the retinal vasculature. Our findings suggest that, through its interaction with YB-1, ZO-1 controls SG formation and the response of ECs to stress during angiogenesis.


Asunto(s)
Células Endoteliales , Proteína 1 de Unión a la Caja Y , Proteína de la Zonula Occludens-1 , Animales , Proteína 1 de Unión a la Caja Y/metabolismo , Proteína 1 de Unión a la Caja Y/genética , Proteína de la Zonula Occludens-1/metabolismo , Proteína de la Zonula Occludens-1/genética , Ratones , Humanos , Células Endoteliales/metabolismo , Gránulos de Estrés/metabolismo , Neovascularización Fisiológica , Vasos Retinianos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Angiogénesis , Factores de Transcripción
18.
Nat Med ; 30(2): 443-454, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38321220

RESUMEN

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 .


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Edema Macular , Animales , Ratones , Humanos , Edema Macular/tratamiento farmacológico , Edema Macular/etiología , Retinopatía Diabética/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Células Endoteliales , Senoterapéuticos , Senescencia Celular
19.
Angiogenesis ; 16(4): 985-92, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23912262

RESUMEN

Regeneration of blood vessels in ischemic neuronal tissue is critical to reduce tissue damage in diseases. In proliferative retinopathy, initial vessel loss leads to retinal ischemia, which can induce either regrowth of vessels to restore normal metabolism and minimize damage, or progress to hypoxia-induced sight-threatening pathologic vaso-proliferation. It is not well understood how retinal neurons mediate regeneration of vascular growth in response to ischemic insults. In this study we aim to investigate the potential role of Sirtuin 1 (Sirt1), a metabolically-regulated protein deacetylase, in mediating the response of ischemic neurons to regulate vascular regrowth in a mouse model of oxygen-induced ischemic retinopathy (OIR). We found that Sirt1 is highly induced in the avascular ischemic retina in OIR. Conditional depletion of neuronal Sirt1 leads to significantly decreased retinal vascular regeneration into the avascular zone and increased hypoxia-induced pathologic vascular growth. This effect is likely independent of PGC-1α, a known Sirt1 target, as absence of PGC-1α in knockout mice does not impact vascular growth in retinopathy. We found that neuronal Sirt1 controls vascular regrowth in part through modulating deacetylation and stability of hypoxia-induced factor 1α and 2α, and thereby modulating expression of angiogenic factors. These results indicate that ischemic neurons induce Sirt1 to promote revascularization into ischemic neuronal areas, suggesting a novel role of neuronal Sirt1 in mediating vascular regeneration in ischemic conditions, with potential implications beyond retinopathy.


Asunto(s)
Isquemia/fisiopatología , Neovascularización Fisiológica/fisiología , Neuronas/metabolismo , Regeneración/fisiología , Vasos Retinianos/fisiología , Retinopatía de la Prematuridad , Sirtuina 1/fisiología , Proteínas Angiogénicas/biosíntesis , Proteínas Angiogénicas/genética , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carbazoles/farmacología , Línea Celular , Modelos Animales de Enfermedad , Isquemia/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Oxígeno/toxicidad , Terapia por Inhalación de Oxígeno/efectos adversos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/enzimología , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/deficiencia , Sirtuina 1/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Regulación hacia Arriba
20.
Blood ; 117(22): 6024-35, 2011 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-21355092

RESUMEN

The failure of blood vessels to revascularize ischemic neural tissue represents a significant challenge for vascular biology. Examples include proliferative retinopathies (PRs) such as retinopathy of prematurity and proliferative diabetic retinopathy, which are the leading causes of blindness in children and working-age adults. PRs are characterized by initial microvascular degeneration, followed by a compensatory albeit pathologic hypervascularization mounted by the hypoxic retina attempting to reinstate metabolic equilibrium. Paradoxically, this secondary revascularization fails to grow into the most ischemic regions of the retina. Instead, the new vessels are misdirected toward the vitreous, suggesting that vasorepulsive forces operate in the avascular hypoxic retina. In the present study, we demonstrate that the neuronal guidance cue semaphorin 3A (Sema3A) is secreted by hypoxic neurons in the avascular retina in response to the proinflammatory cytokine IL-1ß. Sema3A contributes to vascular decay and later forms a chemical barrier that repels neo-vessels toward the vitreous. Conversely, silencing Sema3A expression enhances normal vascular regeneration within the ischemic retina, thereby diminishing aberrant neovascularization and preserving neuroretinal function. Overcoming the chemical barrier (Sema3A) released by ischemic neurons accelerates the vascular regeneration of neural tissues, which restores metabolic supply and improves retinal function. Our findings may be applicable to other neurovascular ischemic conditions such as stroke.


Asunto(s)
Isquemia/patología , Neovascularización Patológica , Neuronas/patología , Oxígeno/toxicidad , Regeneración , Enfermedades de la Retina/patología , Semaforina-3A/fisiología , Animales , Aorta/citología , Aorta/efectos de los fármacos , Aorta/metabolismo , Western Blotting , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Técnicas para Inmunoenzimas , Interleucina-1beta/farmacología , Isquemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , ARN Mensajero/genética , Ratas , Enfermedades de la Retina/etiología , Enfermedades de la Retina/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Neovascularización Retiniana , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA