Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.159
Filtrar
1.
PLoS Pathog ; 20(4): e1012156, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38598560

RESUMEN

SARS-CoV-2 has been shown to cause wide-ranging ocular abnormalities and vision impairment in COVID-19 patients. However, there is limited understanding of SARS-CoV-2 in ocular transmission, tropism, and associated pathologies. The presence of viral RNA in corneal/conjunctival tissue and tears, along with the evidence of viral entry receptors on the ocular surface, has led to speculation that the eye may serve as a potential route of SARS-CoV-2 transmission. Here, we investigated the interaction of SARS-CoV-2 with cells lining the blood-retinal barrier (BRB) and the role of the eye in its transmission and tropism. The results from our study suggest that SARS-CoV-2 ocular exposure does not cause lung infection and moribund illness in K18-hACE2 mice despite the extended presence of viral remnants in various ocular tissues. In contrast, intranasal exposure not only resulted in SARS-CoV-2 spike (S) protein presence in different ocular tissues but also induces a hyperinflammatory immune response in the retina. Additionally, the long-term exposure to viral S-protein caused microaneurysm, retinal pigmented epithelium (RPE) mottling, retinal atrophy, and vein occlusion in mouse eyes. Notably, cells lining the BRB, the outer barrier, RPE, and the inner barrier, retinal vascular endothelium, were highly permissive to SARS-CoV-2 replication. Unexpectedly, primary human corneal epithelial cells were comparatively resistant to SARS-CoV-2 infection. The cells lining the BRB showed induced expression of viral entry receptors and increased susceptibility towards SARS-CoV-2-induced cell death. Furthermore, hyperglycemic conditions enhanced the viral entry receptor expression, infectivity, and susceptibility of SARS-CoV-2-induced cell death in the BRB cells, confirming the reported heightened pathological manifestations in comorbid populations. Collectively, our study provides the first evidence of SARS-CoV-2 ocular tropism via cells lining the BRB and that the virus can infect the retina via systemic permeation and induce retinal inflammation.


Asunto(s)
Barrera Hematorretinal , COVID-19 , Retina , SARS-CoV-2 , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Animales , Barrera Hematorretinal/virología , COVID-19/inmunología , COVID-19/virología , Ratones , Humanos , Retina/virología , Retina/inmunología , Retina/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Internalización del Virus , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/inmunología , Inflamación/inmunología , Inflamación/virología , Betacoronavirus/fisiología , Tropismo Viral , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/patología
2.
Front Immunol ; 15: 1374617, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38665911

RESUMEN

Blindness or vision loss due to neuroretinal and photoreceptor degeneration affects millions of individuals worldwide. In numerous neurodegenerative diseases, including age-related macular degeneration, dysregulated immune response-mediated retinal degeneration has been found to play a critical role in the disease pathogenesis. To better understand the pathogenic mechanisms underlying the retinal degeneration, we used a mouse model of systemic immune activation where we infected mice with lymphocytic choriomeningitis virus (LCMV) clone 13. Here, we evaluated the effects of LCMV infection and present a comprehensive discovery-based proteomic investigation using tandem mass tag (TMT) labeling and high-resolution liquid chromatography-tandem mass spectrometry (LC-MS/MS). Changes in protein regulation in the posterior part of the eye, neuroretina, and RPE/choroid were compared to those in the spleen as a secondary lymphoid organ and to the kidney as a non-lymphoid but encapsulated organ at 1, 8, and 28 weeks of infection. Using bioinformatic tools, we found several proteins responsible for maintaining normal tissue homeostasis to be differentially regulated in the neuroretina and the RPE/choroid during the degenerative process. Additionally, in the organs we observed, several important protein pathways contributing to cellular homeostasis and tissue development were perturbed and associated with LCMV-mediated inflammation, promoting disease progression. Our findings suggest that the response to a systemic chronic infection differs between the neuroretina and the RPE/choroid, and the processes induced by chronic systemic infection in the RPE/choroid are not unlike those induced in non-immune-privileged organs such as the kidney and spleen. Overall, our data provide detailed insight into several molecular mechanisms of neuroretinal degeneration and highlight various novel protein pathways that further suggest that the posterior part of the eye is not an isolated immunological entity despite the existence of neuroretinal immune privilege.


Asunto(s)
Modelos Animales de Enfermedad , Virus de la Coriomeningitis Linfocítica , Proteómica , Degeneración Retiniana , Animales , Ratones , Proteómica/métodos , Degeneración Retiniana/inmunología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones Endogámicos C57BL , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Espectrometría de Masas en Tándem , Proteoma , Retina/inmunología , Retina/metabolismo , Retina/patología , Cromatografía Liquida , Coroides/inmunología , Coroides/patología , Coroides/metabolismo
3.
Int Immunopharmacol ; 133: 112021, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38626549

RESUMEN

BACKGROUND: Diabetic retinopathy (DR) stands as a prevalent secondary complication of diabetes, notably Type 1 Diabetes Mellitus (T1D), characterized by immune system involvement potentially impacting the retinal immune response mediated by microglia. Early stages of DR witness blood-retinal barrier permeabilization, facilitating peripheral immune cell interaction with the retinal immune system. Kaempferol (Kae), known for its potent anti-inflammatory activity, presents a promising avenue in DR treatment by targeting the immune mechanisms underlying its onset and progression. Our investigation delves into the molecular intricacies of innate immune cell interaction during DR progression and the attenuation of inflammatory processes pivotal to its pathology. METHODS: Employing in vitro studies, we exposed HAPI microglial and J774.A1 macrophage cells to pro-inflammatory stimuli in the presence or absence of Kae. Ex vivo and in vivo experiments utilized BB rats, a T1D animal model. Retinal explants from BB rats were cultured with Kae, while intraperitoneal Kae injections were administered to BB rats for 15 days. Quantitative PCR, Western blotting, immunofluorescence, and Spectral Domain - Optical Coherence Tomography (SD-OCT) facilitated survival assessment, cellular signaling analysis, and inflammatory marker determination. RESULTS: Results demonstrate Kae significantly mitigates inflammatory processes across in vitro, ex vivo, and in vivo DR models, primarily targeting immune cell responses. Kae administration notably inhibits proinflammatory responses during DR progression while promoting an anti-inflammatory milieu, chiefly through microglia-mediated synthesis of Arginase-1 and Hemeoxygenase-1(HO-1). In vivo, Kae administration effectively preserves retinal integrity amid DR progression. CONCLUSIONS: Our findings elucidate the interplay between retinal and systemic immune cells in DR progression, underscoring a differential treatment response predominantly orchestrated by microglia's anti-inflammatory action. Kae treatment induces a phenotypic and functional shift in immune cells, delaying DR progression, thereby spotlighting microglial cells as a promising therapeutic target in DR management.


Asunto(s)
Retinopatía Diabética , Quempferoles , Macrófagos , Microglía , Animales , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/inmunología , Retinopatía Diabética/patología , Microglía/efectos de los fármacos , Microglía/inmunología , Quempferoles/farmacología , Quempferoles/uso terapéutico , Ratas , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Progresión de la Enfermedad , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/inmunología , Retina/efectos de los fármacos , Retina/patología , Retina/inmunología , Línea Celular , Masculino , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Humanos , Agentes Inmunomoduladores/farmacología , Agentes Inmunomoduladores/uso terapéutico , Modelos Animales de Enfermedad
4.
J Mol Med (Berl) ; 102(5): 585-597, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38429382

RESUMEN

Diabetic retinopathy (DR) is recognized as a neurovascular complication of diabetes, and emerging evidence underscores the pivotal role of inflammation in its pathophysiology. Macrophage activation is increasingly acknowledged as a key contributor to the onset and progression of DR. Different populations of macrophages originating from distinct sources contribute to DR-associated inflammation. Retinal macrophages can be broadly categorized into two main groups based on their origin: intrinsic macrophages situated within the retina and vitreoretinal interface and macrophages derived from infiltrating monocytes. The former comprises microglia (MG), perivascular macrophages, and macrophage-like hyalocytes. Retinal MG, as the principal population of tissue-resident population of mononuclear phagocytes, exhibits high heterogeneity and plasticity while serving as a crucial connector between retinal capillaries and synapses. This makes MG actively involved in the pathological processes across various stages of DR. Activated hyalocytes also contribute to the pathological progression of advanced DR. Additionally, recruited monocytes, displaying rapid turnover in circulation, augment the population of retinal macrophages during DR pathogenesis, exerting pathogenic or protective effect based on different subtypes. In this review, we examine novel perspectives on macrophage biology based on recent studies elucidating the diversity of macrophage identity and function, as well as the mechanisms influencing macrophage behavior. These insights may pave the way for innovative therapeutic strategies in the management of DR.


Asunto(s)
Retinopatía Diabética , Activación de Macrófagos , Macrófagos , Retinopatía Diabética/inmunología , Retinopatía Diabética/patología , Humanos , Animales , Macrófagos/inmunología , Macrófagos/metabolismo , Activación de Macrófagos/inmunología , Retina/patología , Retina/inmunología , Retina/metabolismo , Microglía/inmunología , Microglía/patología , Microglía/metabolismo , Monocitos/inmunología , Monocitos/metabolismo
5.
Vis Neurosci ; 39: E005, 2022 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-36164752

RESUMEN

To study the macroglia and microglia and the immune role in long-time light exposure in rat eyes, we performed glial cell characterization along the time-course of retinal degeneration induced by chronic exposure to low-intensity light. Animals were exposed to light for periods of 2, 4, 6, or 8 days, and the retinal glial response was evaluated by immunohistochemistry, western blot and real-time reverse transcription polymerase chain reaction. Retinal cells presented an increased expression of the macroglia marker GFAP, as well as increased mRNA levels of microglia markers Iba1 and CD68 after 6 days. Also, at this time-point, we found a higher number of Iba1-positive cells in the outer nuclear layer area; moreover, these cells showed the characteristic activated-microglia morphology. The expression levels of immune mediators TNF, IL-6, and chemokines CX3CR1 and CCL2 were also significantly increased after 6 days. All the events of glial activation occurred after 5-6 days of constant light exposure, when the number of photoreceptor cells has already decreased significantly. Herein, we demonstrated that glial and immune activation are secondary to neurodegeneration; in this scenario, our results suggest that photoreceptor death is an early event that occurs independently of glial-derived immune responses.


Asunto(s)
Interleucina-6 , Neuroglía , Traumatismos por Radiación , Retina , Degeneración Retiniana , Animales , Quimiocinas/genética , Quimiocinas/metabolismo , Interleucina-6/metabolismo , Luz , Neuroglía/inmunología , ARN Mensajero/genética , Traumatismos por Radiación/etiología , Traumatismos por Radiación/inmunología , Ratas , Retina/inmunología , Retina/efectos de la radiación , Degeneración Retiniana/etiología , Degeneración Retiniana/inmunología
6.
Biochem Pharmacol ; 197: 114917, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35041813

RESUMEN

Noninfectious (autoimmune and immune-mediated) uveitis is one of the primary diseases leading to blindness in the world. Due to the limitation of current first-line drugs for clinical uveitis, novel drugs and targets against uveitis are urgently needed. Ganciclovir (GCV), an FDA-approved antiviral drug, is often used to treat cytomegalovirus-induced retinitis in clinical patients. Recently, GCV was found to suppress neuroinflammation via targeting STING signaling because the STING pathway plays a pivotal role in autoimmune diseases. However, until now, the effect of GCV on non-infectious uveitis has never been explored. In this work, using the rat experimental autoimmune uveitis (EAU) model, we first found STING to be highly expressed in infiltrating cells (CD68+, CD45+, and CD4+) and retinal glial cells (Iba1+ and GFAP+) of the immunized retina. More importantly, GCV treatment can significantly suppress the initiation and progression of EAU by inhibiting infiltration of Th17 and inflammatory cells into the retina. Mechanistically, we found that GCV could reverse the levels of pro-inflammatory factors (such as IL-1ß) and chemokine-related factors (such as Cxcr3), possibly via targeting the STING pathway. The present results suggest that GCV may be considered as a novel therapeutic strategy against human uveitis.


Asunto(s)
Enfermedades Autoinmunes/prevención & control , Ganciclovir/uso terapéutico , Mediadores de Inflamación/antagonistas & inhibidores , Retina/efectos de los fármacos , Células Th17/efectos de los fármacos , Uveítis/prevención & control , Animales , Enfermedades Autoinmunes/inducido químicamente , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Proteínas del Ojo/toxicidad , Ganciclovir/farmacología , Humanos , Mediadores de Inflamación/inmunología , Masculino , Ratas , Ratas Endogámicas Lew , Retina/inmunología , Retina/patología , Proteínas de Unión al Retinol/toxicidad , Células Th17/inmunología , Células Th17/patología , Uveítis/inducido químicamente , Uveítis/inmunología , Uveítis/patología
7.
Cells ; 10(12)2021 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-34944083

RESUMEN

Considering the fact that many retinal diseases are yet to be cured, the pathomechanisms of these multifactorial diseases need to be investigated in more detail. Among others, oxidative stress and hypoxia are pathomechanisms that take place in retinal diseases, such as glaucoma, age-related macular degeneration, or diabetic retinopathy. In consideration of these diseases, it is also evidenced that the immune system, including the complement system and its activation, plays an important role. Suitable models to investigate neuroretinal diseases are organ cultures of porcine retina. Based on an established model, the role of the complement system was studied after the induction of oxidative stress or hypoxia. Both stressors led to a loss of retinal ganglion cells (RGCs) accompanied by apoptosis. Hypoxia activated the complement system as noted by higher C3+ and MAC+ cell numbers. In this model, activation of the complement cascade occurred via the classical pathway and the number of C1q+ microglia was increased. In oxidative stressed retinas, the complement system had no consideration, but strong inflammation took place, with elevated TNF, IL6, and IL8 mRNA expression levels. Together, this study shows that hypoxia and oxidative stress induce different mechanisms in the porcine retina inducing either the immune response or an inflammation. Our findings support the thesis that the immune system is involved in the development of retinal diseases. Furthermore, this study is evidence that both approaches seem suitable models to investigate undergoing pathomechanisms of several neuroretinal diseases.


Asunto(s)
Activación de Complemento/inmunología , Vía Clásica del Complemento/inmunología , Hipoxia/inmunología , Retina/inmunología , Retina/patología , Células Ganglionares de la Retina/patología , Animales , Apoptosis/efectos de los fármacos , Cobalto/toxicidad , Activación de Complemento/efectos de los fármacos , Vía Alternativa del Complemento/efectos de los fármacos , Vía Alternativa del Complemento/inmunología , Vía Clásica del Complemento/efectos de los fármacos , Proteínas del Sistema Complemento/metabolismo , Peróxido de Hidrógeno/toxicidad , Lectinas/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Estrés Oxidativo/efectos de los fármacos , Células Ganglionares de la Retina/efectos de los fármacos , Neuronas Retinianas/efectos de los fármacos , Neuronas Retinianas/patología , Estrés Fisiológico/efectos de los fármacos , Porcinos
8.
Invest Ophthalmol Vis Sci ; 62(15): 31, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34967854

RESUMEN

Purpose: The purpose of this study was to elucidate the effects of interleukin (IL)-38 on experimental autoimmune uveitis (EAU) and its underlying mechanisms. Methods: Mice with EAU were treated with IL-38, and the retinas and cervical draining lymph nodes (CDLNs) were analyzed by flow cytometry. Single-cell RNA sequencing (scRNA-seq) was conducted to analyze the immune cell profiles of CDLNs from normal, EAU, and IL-38-treated mice. Results: Administration of IL-38 attenuated EAU symptoms and reduced the proportion of T helper 17 (Th17) and T helper 1 (Th1) cells in the retinas and CDLNs. In scRNA-seq analysis, IL-38 downregulated the IL-17 signaling pathway and reduced the expression of Th17 cell pathogenicity-related genes (Csf2 and Il23r), findings which were also confirmed by flow cytometry. In vitro, IL-38 reduced the granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulation function of IL-23 and inhibited IL-23R expression in Th17 cells. Moreover, when co-cultured with Th17 cells, IL-38 prevented IL-23 production in antigen-presenting cells (APCs). Conclusions: Our data demonstrate the therapeutic effect of IL-38 on EAU, and suggest that the effect of IL-38 may be caused by dampening of the GM-CSF/IL-23R/IL-23 feedback loop between Th17 cells and APCs.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Sistema Inmunológico/fisiología , Interleucinas/uso terapéutico , Células Th17/inmunología , Uveítis/tratamiento farmacológico , Traslado Adoptivo , Animales , Células Presentadoras de Antígenos/inmunología , Enfermedades Autoinmunes/inducido químicamente , Enfermedades Autoinmunes/inmunología , Linfocitos B/inmunología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inyecciones Intravenosas , Interleucina-23/metabolismo , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Cuello , Proteínas Recombinantes/uso terapéutico , Retina/inmunología , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Linfocitos T/inmunología , Células TH1/inmunología , Uveítis/inducido químicamente , Uveítis/inmunología
9.
Cells ; 10(11)2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34831487

RESUMEN

Diabetic retinopathy is a frequent complication of longstanding diabetes, which comprises a complex interplay of microvascular abnormalities and neurodegeneration. Zebrafish harboring a homozygous mutation in the pancreatic transcription factor pdx1 display a diabetic phenotype with survival into adulthood, and are therefore uniquely suitable among zebrafish models for studying pathologies associated with persistent diabetic conditions. We have previously shown that, starting at three months of age, pdx1 mutants exhibit not only vascular but also neuro-retinal pathologies manifesting as photoreceptor dysfunction and loss, similar to human diabetic retinopathy. Here, we further characterize injury and regenerative responses and examine the effects on progenitor cell populations. Consistent with a negative impact of hyperglycemia on neurogenesis, stem cells of the ciliary marginal zone show an exacerbation of aging-related proliferative decline. In contrast to the robust Müller glial cell proliferation seen following acute retinal injury, the pdx1 mutant shows replenishment of both rod and cone photoreceptors from slow-cycling, neurod-expressing progenitors which first accumulate in the inner nuclear layer. Overall, we demonstrate a diabetic retinopathy model which shows pathological features of the human disease evolving alongside an ongoing restorative process that replaces lost photoreceptors, at the same time suggesting an unappreciated phenotypic continuum between multipotent and photoreceptor-committed progenitors.


Asunto(s)
Hiperglucemia/patología , Células-Madre Neurales/patología , Retina/patología , Envejecimiento/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Muerte Celular , Proliferación Celular , Enfermedad Crónica , Células Ependimogliales/patología , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Modelos Biológicos , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción PAX6/metabolismo , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Receptores Notch/metabolismo , Retina/inmunología , Transducción de Señal , Transactivadores/genética , Pez Cebra
10.
Front Immunol ; 12: 761776, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34745138

RESUMEN

Confocal scanning laser ophthalmoscopy (cSLO) is a non-invasive technique for real-time imaging of the retina. We developed a step-by-step protocol for the semi-automatic evaluation of myeloid cells in cSLO images from CX3CR1GFP mice, expressing green fluorescent protein (GFP) under control of the endogenous CX3C chemokine receptor 1 locus. We identified cSLO parameters allowing us to distinguish animals with experimental autoimmune encephalomyelitis (EAE) from sham-treated/naïve animals. Especially cell count (CC) and the total microglial area (SuA) turned out to be reliable parameters. Comparing the cSLO results with clinical parameters, we found significant correlations between the clinical EAE score and the SuA and of the inner retinal layer thickness, measured by optical coherence tomography, with the CC as well as the SuA. As a final step, we performed immunohistochemistry to confirm that the GFP-expressing cells visualized by the cSLO are Iba1 positive and validated the step-by-step protocol against manual counting. We present a semi-automatic step-by-step protocol with a balance between fast data evaluation and adequate accuracy, which is optimized by the option to manually adapt the contrast threshold. This protocol may be useful for numerous research questions on the role of microglial polarization in models of inflammatory and degenerating CNS diseases involving the retina.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Microglía/inmunología , Animales , Receptor 1 de Quimiocinas CX3C/genética , Encefalomielitis Autoinmune Experimental/diagnóstico por imagen , Femenino , Proteínas Fluorescentes Verdes/genética , Masculino , Ratones Transgénicos , Oftalmoscopía/métodos , Retina/diagnóstico por imagen , Retina/inmunología , Tomografía de Coherencia Óptica
11.
J Immunol Res ; 2021: 2389029, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34692851

RESUMEN

Diabetic retinopathy (DR), as a major cause of blindness worldwide, is one common complication of diabetes mellitus. Inflammatory response and oxidative stress injury of endothelial cells play significant roles in the pathogenesis of DR. The study is aimed at investigating the effects of lysophosphatidylcholine (LPC) on the dysfunction of high glucose- (HG-) treated human retinal microvascular endothelial cells (HRMECs) after being cocultured with bone marrow mesenchymal stem cells (BMSCs) and the underlying regulatory mechanism. Coculture of BMSCs and HRMECs was performed in transwell chambers. The activities of antioxidant-related enzymes and molecules of oxidative stress injury and the contents of inflammatory cytokines were measured by ELISA. Flow cytometry analyzed the apoptosis of treated HRMECs. HRMECs were further treated with 10-50 µg/ml LPC to investigate the effect of LPC on the dysfunction of HRMECs. Western blotting was conducted to evaluate levels of TLR4 and p-NF-κB proteins. We found that BMSCs alleviated HG-induced inflammatory response and oxidative stress injury of HRMECs. Importantly, LPC offsets the protective effects of BMSCs on inflammatory response and oxidative stress injury of HRMECs. Furthermore, LPC upregulated the protein levels of TLR4 and p-NF-κB, activating the TLR4/NF-κB signaling pathway. Overall, our study demonstrated that LPC offsets the protective effects of BMSCs on inflammatory response and oxidative stress injury of HRMECs via TLR4/NF-κB signaling.


Asunto(s)
Retinopatía Diabética/terapia , Lisofosfatidilcolinas/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Células Cultivadas , Técnicas de Cocultivo , Retinopatía Diabética/inmunología , Células Endoteliales/inmunología , Células Endoteliales/patología , Humanos , FN-kappa B/metabolismo , Estrés Oxidativo/inmunología , Cultivo Primario de Células , Retina/citología , Retina/inmunología , Retina/patología , Transducción de Señal/inmunología , Receptor Toll-Like 4/metabolismo
13.
Front Immunol ; 12: 724601, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34484232

RESUMEN

The ocular tissue microenvironment is immune privileged and uses several mechanisms of immunosuppression to prevent the induction of inflammation. Besides being a blood-barrier and source of photoreceptor nutrients, the retinal pigment epithelial cells (RPE) regulate the activity of immune cells within the retina. These mechanisms involve the expression of immunomodulating molecules that make macrophages and microglial cells suppress inflammation and promote immune tolerance. The RPE have an important role in ocular immune privilege to regulate the behavior of immune cells within the retina. Reviewed is the current understanding of how RPE mediate this regulation and the changes seen under pathological conditions.


Asunto(s)
Macrófagos/inmunología , Microglía/inmunología , Retina/trasplante , Epitelio Pigmentado de la Retina/trasplante , Animales , Humanos , Tolerancia Inmunológica , Ratones , Retina/inmunología , Epitelio Pigmentado de la Retina/inmunología , Inmunología del Trasplante , Trasplante Homólogo
14.
Cells ; 10(7)2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34359839

RESUMEN

Neuroinflammation has been put forward as a mechanism triggering axonal regrowth in the mammalian central nervous system (CNS), yet little is known about the underlying cellular and molecular players connecting these two processes. In this study, we provide evidence that MMP2 is an essential factor linking inflammation to axonal regeneration by using an in vivo mouse model of inflammation-induced axonal regeneration in the optic nerve. We show that infiltrating myeloid cells abundantly express MMP2 and that MMP2 deficiency results in reduced long-distance axonal regeneration. However, this phenotype can be rescued by restoring MMP2 expression in myeloid cells via a heterologous bone marrow transplantation. Furthermore, while MMP2 deficiency does not affect the number of infiltrating myeloid cells, it does determine the coordinated expression of pro- and anti-inflammatory molecules. Altogether, in addition to its role in axonal regeneration via resolution of the glial scar, here, we reveal a new mechanism via which MMP2 facilitates axonal regeneration, namely orchestrating the expression of pro- and anti-inflammatory molecules by infiltrating innate immune cells.


Asunto(s)
Axones/inmunología , Trasplante de Médula Ósea , Metaloproteinasa 2 de la Matriz/genética , Regeneración Nerviosa/inmunología , Traumatismos del Nervio Óptico/inmunología , Nervio Óptico/inmunología , Animales , Antígenos Ly/genética , Antígenos Ly/inmunología , Axones/ultraestructura , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/inmunología , Movimiento Celular , Proteína GAP-43/genética , Proteína GAP-43/inmunología , Regulación de la Expresión Génica , Inmunidad Innata , Inflamación , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/inmunología , Metaloproteinasa 2 de la Matriz/deficiencia , Metaloproteinasa 2 de la Matriz/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/citología , Células Mieloides/inmunología , Regeneración Nerviosa/genética , Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/genética , Traumatismos del Nervio Óptico/patología , Retina/inmunología , Retina/lesiones , Retina/metabolismo , Trasplante Heterólogo , Irradiación Corporal Total
15.
Sci Rep ; 11(1): 17254, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34446787

RESUMEN

Age-related macular degeneration (AMD) is genetically associated with complement. Dendritic cells (DCs) play key roles during innate and adaptive immunity, and express complement components and their receptors. We investigated ocular DC heterogeneity and the role of DCs in the laser-induced choroidal neovascularization (CNV) model. In order to determine the function of DCs, we used two models of DC deficiency: the Flt3-/- and Flt3l-/- mouse. We identified three types of ocular DCs: plasmacytoid DC, classical DC-1, and classical DC-2. At steady-state, classical DCs were found in the iris and choroid but were not detectable in the retina. Plasmacytoid DCs existed at very low levels in iris, choroid, and retina. After laser injury, the number of each DC subset was up-regulated in the choroid and retina. In Flt3-/- mice, we found reduced numbers of classical DCs at steady-state, but each DC subset equally increased after laser injury between wildtype and Flt3-/- mice. In Flt3l-/- mice, each DC subsets was severely reduced after laser injury. Neither Flt3-/- or Flt3l-/- mice demonstrated reduced CNV area compared to wildtype mice. DCs do not play any significant role during the laser-induced CNV model of neovascular AMD.


Asunto(s)
Coroides/inmunología , Neovascularización Coroidal/inmunología , Células Dendríticas/inmunología , Proteínas de la Membrana/inmunología , Tirosina Quinasa 3 Similar a fms/inmunología , Animales , Coroides/irrigación sanguínea , Neovascularización Coroidal/etiología , Neovascularización Coroidal/genética , Femenino , Citometría de Flujo/métodos , Iris/irrigación sanguínea , Iris/inmunología , Rayos Láser/efectos adversos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Retina/inmunología , Agudeza Visual/inmunología , Degeneración Macular Húmeda/inmunología , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo
16.
Biomolecules ; 11(8)2021 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-34439829

RESUMEN

Retinitis pigmentosa (RP) is a hereditary disease of the retina that results in complete blindness. Currently, there are very few treatments for the disease and those that exist work only for the recessively inherited forms. To better understand the pathogenesis of RP, multiple mouse models have been generated bearing mutations found in human patients including the human Q344X rhodopsin knock-in mouse. In recent years, the immune system was shown to play an increasingly important role in RP degeneration. By way of electroretinography, optical coherence tomography, funduscopy, fluorescein angiography, and fluorescent immunohistochemistry, we show degenerative and vascular phenotypes, microglial activation, photoreceptor phagocytosis, and upregulation of proinflammatory pathway proteins in the retinas of the human Q344X rhodopsin knock-in mouse. We also show that an FDA-approved pharmacological agent indicated for the treatment of rheumatoid arthritis is able to halt activation of pro-inflammatory signaling in cultured retinal cells, setting the stage for pre-clinical trials using these mice to inhibit proinflammatory signaling in an attempt to preserve vision. We conclude from this work that pro- and autoinflammatory upregulation likely act to enhance the progression of the degenerative phenotype of rhodopsin Q344X-mediated RP and that inhibition of these pathways may lead to longer-lasting vision in not only the Q344X rhodopsin knock-in mice, but humans as well.


Asunto(s)
Antirreumáticos/farmacología , Compuestos Heterocíclicos con 3 Anillos/farmacología , Factor Inhibidor de Leucemia/farmacología , Mutación , Retina/efectos de los fármacos , Retinitis Pigmentosa/tratamiento farmacológico , Rodopsina/genética , Sustitución de Aminoácidos , Animales , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Expresión Génica , Técnicas de Sustitución del Gen , Humanos , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/genética , Quinasas Janus/inmunología , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/patología , FN-kappa B/genética , FN-kappa B/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Retina/inmunología , Retina/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/inmunología , Retinitis Pigmentosa/patología , Rodopsina/deficiencia , Factores de Transcripción STAT/antagonistas & inhibidores , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/inmunología , Transducción de Señal , Transgenes , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
17.
Invest Ophthalmol Vis Sci ; 62(10): 10, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34379096

RESUMEN

Purpose: In spite of clear differences in tissue function and significance to ocular disease, little is known about how immune responses differ between the retina and uveal tract. To this end we compared the effects of acute systemic inflammation on myeloid cells within the mouse retina, iris-ciliary body, and choroid. Methods: Systemic inflammation was induced in Cx3cr1gfp/gfp and CD11c-eYFP Crb1wt/wtmice by intraperitoneal lipopolysaccharide (LPS). In vivo fundus imaging was performed at two, 24, and 48 hours after LPS, and ocular tissue wholemounts were immunostained and studied by confocal microscopy. Flow cytometry was used to investigate the expression of activation markers (MHC class II, CD80, CD86) on myeloid cell populations at 24 hours. For functional studies, retinal microglia were isolated from LPS-exposed mice and cocultured with naïve OT-II CD4+ T-cells and ovalbumin peptide. T-cell proliferation was measured by flow cytometry and cytokine assays. Results: Systemic LPS altered the density and morphology of retinal microglia; however, retinal microglia did not upregulate antigen presentation markers and failed to stimulate naïve CD4+ T-cell proliferation in vitro. In contrast, uveal tract myeloid cells displayed a phenotype consistent with late-activated antigen-presenting cells at 24 hours. Systemic LPS induced remodeling of myeloid populations within the uveal tract, particularly in the choroid, where dendritic cells were partially displaced by macrophages at 24 hours. Conclusions: The disparate myeloid cell responses in the retina and uveal tract after systemic LPS highlight differential regulation of innate immunity within these tissue environments, observations that underpin and advance our understanding of ocular immune privilege.


Asunto(s)
Células Dendríticas/patología , Inflamación/patología , Macrófagos/patología , Células Mieloides/patología , Retina/patología , Úvea/patología , Animales , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Inflamación/inmunología , Inflamación/metabolismo , Macrófagos/inmunología , Ratones Endogámicos BALB C , Microscopía Confocal , Células Mieloides/inmunología , Retina/inmunología , Úvea/inmunología
18.
Sci Rep ; 11(1): 15047, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34294798

RESUMEN

Autoimmune retinopathy (AIR) is a rare immune-mediated inflammation of the retina. The autoantibodies against retinal proteins and glycolytic enzymes were reported to be involved in the pathogenesis. This retrospective cohort study assessed the antiretinal autoantibody profiles and their association with clinical outcomes of AIR patients in Thailand. We included 44 patients, 75% were females, with the overall median age of onset of 48 (17-74, IQR 40-55.5) years. Common clinical presentations were nyctalopia (65.9%), blurred vision (52.3%), constricted visual field (43.2%), and nonrecordable electroretinography (65.9%). Underlying malignancy and autoimmune diseases were found in 2 and 12 female patients, respectively. We found 41 autoantibodies, with anti-α-enolase (65.9%) showing the highest prevalence, followed by anti-CAII (43.2%), anti-aldolase (40.9%), and anti-GAPDH (36.4%). Anti-aldolase was associated with male gender (P = 0.012, OR 7.11, 95% CI 1.54-32.91). Anti-CAII showed significant association with age of onset (P = 0.025, 95% CI - 17.28 to - 1.24), while anti-α-enolase (P = 0.002, OR 4.37, 95% CI 1.83-10.37) and anti-GAPDH (P = 0.001, OR 1.87, 95% CI 1.32-2.64) were significantly associated with nonrecordable electroretinography. Association between the antibody profiles and clinical outcomes may be used to direct and adjust the treatment plans and provide insights in the pathogenesis of AIR.


Asunto(s)
Autoanticuerpos/inmunología , Enfermedades Autoinmunes/epidemiología , Enfermedades Autoinmunes/inmunología , Autoinmunidad , Susceptibilidad a Enfermedades , Enfermedades de la Retina/epidemiología , Enfermedades de la Retina/inmunología , Adolescente , Adulto , Anciano , Autoanticuerpos/sangre , Autoantígenos/inmunología , Enfermedades Autoinmunes/diagnóstico , Biomarcadores , Susceptibilidad a Enfermedades/inmunología , Electrorretinografía , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Retina/inmunología , Enfermedades de la Retina/diagnóstico , Estudios Retrospectivos , Adulto Joven
19.
Front Immunol ; 12: 680568, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093583

RESUMEN

Autoimmune uveitis is a sight-threatening ocular inflammatory condition in which the retina and uveal tissues become a target of autoreactive immune cells. The CD47 is a ubiquitously expressed transmembrane protein which plays multiple roles in fundamental cellular functions including phagocytosis, proliferation, and adhesion. Signal regulatory protein alpha (SIRPα), one of the CD47 ligands, is predominantly expressed in myeloid lineage cells such as dendritic cells (DCs) or macrophages, and CD47-SIRPα signaling pathway is implicated in the development of autoimmune diseases. Our current study demonstrates how CD47 depletion is effective in the prevention of experimental autoimmune uveitis (EAU), an animal model of human autoimmune uveitis, in animals deficient of CD47 (CD47-/- ). Systemic suppression of SIRPα+ DCs in animals deficient in CD47 resulted in the inability of autoreactive CD4+ T cells to develop, which is crucial to induction of EAU. Of interest, retinal microglia, the resident immune cell of the retina, express SIRPα, however these cells were not operative in EAU suppression in response to CD47 depletion. These results identify CD47 as a significant regulator in the development of SIRPα+ DCs that is vital to disease induction in EAU.


Asunto(s)
Enfermedades Autoinmunes/etiología , Antígeno CD47/deficiencia , Susceptibilidad a Enfermedades , Oftalmopatías/etiología , Animales , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/metabolismo , Autoinmunidad/genética , Biomarcadores , Modelos Animales de Enfermedad , Oftalmopatías/diagnóstico , Oftalmopatías/metabolismo , Femenino , Inmunomodulación/genética , Inmunofenotipificación , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones Noqueados , Retina/inmunología , Retina/metabolismo , Retina/patología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Uveítis/diagnóstico , Uveítis/etiología , Uveítis/metabolismo
20.
Invest Ophthalmol Vis Sci ; 62(7): 21, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34144609

RESUMEN

Purpose: For this study we aimed to understand if retinal pigment epithelial (RPE) cells express antimicrobial peptide lysozyme as a mechanism to protect the neuroretina from blood-borne pathogens. Methods: The expression of lysozyme in human and mouse RPE cells was examined by RT-PCR or immune (cyto)histochemistry in cell cultures or retinal sections. RPE cultures were treated with different concentrations of Pam3CSK4, lipopolysaccharides (LPS), staphylococcus aureus-derived peptidoglycan (PGN-SA), Poly(I:C), and Poly(dA:dT). The mRNA expression of lysozyme was examined by qPCR and protein expression by ELISA. Poly(I:C) was injected into the subretinal space of C57BL/6J mice and eyes were collected 24 hours later and processed for the evaluation of lysozyme expression by confocal microscopy. Bactericidal activity was measured in ARPE19 cells following LYZ gene deletion using Crispr/Cas9 technology. Results: The mRNA and protein of lysozyme were detected in mouse and human RPE cells under normal conditions, although the expression levels were lower than mouse microglia BV2 or human monocytes THP-1 cells, respectively. Immunohistochemistry showed punctate lysozyme expression inside RPE cells. Lysozyme was detected by ELISA in normal RPE lysates, and in live bacteria-treated RPE supernatants. Treatment of RPE cells with Pam3CSK4, LPS, PGN-SA, and Poly(I:C) enhanced lysozyme expression. CRISPR/Cas9 deletion of lysozyme impaired bactericidal activity of ARPE19 cells and reduced their response to LPS and Poly(I:C) stimulation. Conclusions: RPE cells constitutively express antimicrobial peptide lysozyme and the expression is modulated by pathogenic challenges. RPE cells may protect the neuroretina from blood-borne pathogens by producing antimicrobial peptides, such as lysozyme.


Asunto(s)
Lipopéptidos/fisiología , Muramidasa , Retina , Animales , Antibacterianos/metabolismo , Antibacterianos/farmacología , Barrera Hematorretinal/inmunología , Barrera Hematorretinal/metabolismo , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Ratones , Muramidasa/genética , Muramidasa/farmacología , Poli I-C/metabolismo , Poli I-C/farmacología , Factores Protectores , Retina/inmunología , Retina/metabolismo , Epitelio Pigmentado de la Retina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA