Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.170
Filtrar
1.
PLoS One ; 19(7): e0304073, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38968328

RESUMO

BACKGROUND: Pharmacologic immunosuppression regimes are commonly employed in stem cell clinical trials to mitigate host immune rejection and promote survival and viability of transplanted cells. Immunosuppression and cell survival has been extensively studied in retinal and spinal tissues. The applicability of stem cell therapy is rapidly expanding to other sensory organs such as the ear and hearing. As regenerative therapy is directed to new areas, a greater understanding of immunosuppression strategies and their efficacy is required to facilitate translation to organ-specific biologic microenvironments. OBJECTIVE: This systematic review appraises the current literature regarding immunosuppression strategies employed in stem cell trials of retinal and neural cells. METHODS: This systematic review was performed in line with Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Inclusion criteria included studies presenting data on neural or retinal cells as part of an in-human clinical trial that detailed the immunosuppression regime used. Exclusion criteria included non-English language studies, animal studies, review articles, case reports, editorials, and letters. The databases Medline, Embase, Scopus, Web of Science, and the Cochrane Library were searched from inception to February 2024. Risk of bias was evaluated using the ROBINS-I tool. RESULTS: Eighteen articles fit the inclusion criteria. Nine articles concerned retinal cells, 5 concerned spinal cord injury, and 4 concerned amyotrophic lateral sclerosis. A multi-drug and short-term immunosuppression regime were commonly employed in the identified studies. Detected immune responses in treated patients were rare. Common immunosuppression paradigms included tacrolimus, mycophenolate mofetil and tapering doses of steroids. Local immunosuppression with steroids was employed in some studies concerning retinal diseases. DISCUSSION: A short-term course of systemic immunosuppression seemed efficacious for most included studies, with some showing grafted cells viable months to years after immunosuppression had stopped. Longer-term follow-up is required to see if this remains the case. Side effects related to immunosuppression were uncommon.


Assuntos
Terapia de Imunossupressão , Transplante de Células-Tronco , Humanos , Transplante de Células-Tronco/métodos , Terapia de Imunossupressão/métodos , Retina/imunologia , Imunossupressores/uso terapêutico , Ensaios Clínicos como Assunto
2.
Neurol Neuroimmunol Neuroinflamm ; 11(5): e200273, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38941573

RESUMO

BACKGROUND AND OBJECTIVES: To systematically describe the clinical picture of double-antibody seronegative neuromyelitis optica spectrum disorders (DN-NMOSD) with specific emphasis on retinal involvement. METHODS: Cross-sectional data of 25 people with DN-NMOSD (48 eyes) with and without a history of optic neuritis (ON) were included in this study along with data from 25 people with aquaporin-4 antibody seropositive neuromyelitis optica spectrum disorder (AQP4-NMOSD, 46 eyes) and from 25 healthy controls (HCs, 49 eyes) for comparison. All groups were matched for age and sex and included from the collaborative retrospective study of retinal optical coherence tomography (OCT) in neuromyelitis optica (CROCTINO). Participants underwent OCT with central postprocessing and local neurologic examination and antibody testing. Retinal neurodegeneration was quantified as peripapillary retinal nerve fiber layer thickness (pRNFL) and combined ganglion cell and inner plexiform layer thickness (GCIPL). RESULTS: This DN-NMOSD cohort had a history of [median (inter-quartile range)] 6 (5; 9) attacks within their 5 ± 4 years since onset. Myelitis and ON were the most common attack types. In DN-NMOSD eyes after ON, pRNFL (p < 0.001) and GCIPL (p = 0.023) were thinner compared with eyes of HCs. Even after only one ON episode, DN-NMOSD eyes already had considerable neuroaxonal loss compared with HCs. In DN-NMOSD eyes without a history of ON, pRNFL (p = 0.027) and GCIPL (p = 0.022) were also reduced compared with eyes of HCs. However, there was no difference in pRNFL and GCIPL between DN-NMOSD and AQP4-NMOSD for the whole group and for subsets with a history of ON and without a history of ON-as well as between variances of retinal layer thicknesses. DISCUSSION: DN-NMOSD is characterized by severe retinal damage after ON and attack-independent retinal neurodegeneration. Most of the damage occurs during the first ON episode, which highlights the need for better diagnostic markers in DN-NMOSD to facilitate an earlier diagnosis as well as for effective and early treatments. In this study, people with DN-NMOSD presented with homogeneous clinical and imaging findings potentially suggesting a common retinal pathology in these patients.


Assuntos
Aquaporina 4 , Neuromielite Óptica , Tomografia de Coerência Óptica , Humanos , Neuromielite Óptica/diagnóstico por imagem , Neuromielite Óptica/imunologia , Neuromielite Óptica/sangue , Feminino , Masculino , Adulto , Estudos Transversais , Pessoa de Meia-Idade , Aquaporina 4/imunologia , Estudos Retrospectivos , Autoanticorpos/sangue , Retina/diagnóstico por imagem , Retina/patologia , Retina/imunologia
3.
Medicina (Kaunas) ; 60(6)2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38929562

RESUMO

The complement cascade is a vital system in the human body's defense against pathogens. During the natural aging process, it has been observed that this system is imperative for ensuring the integrity and homeostasis of the retina. While this system is critical for proper host defense and retinal integrity, it has also been found that dysregulation of this system may lead to certain retinal pathologies, including geographic atrophy and diabetic retinopathy. Targeting components of the complement system for retinal diseases has been an area of interest, and in vivo, ex vivo, and clinical trials have been conducted in this area. Following clinical trials, medications targeting the complement system for retinal disease have also become available. In this manuscript, we discuss the pathophysiology of complement dysfunction in the retina and specific pathologies. We then describe the results of cellular, animal, and clinical studies targeting the complement system for retinal diseases. We then provide an overview of complement inhibitors that have been approved by the Food and Drug Administration (FDA) for geographic atrophy. The complement system in retinal diseases continues to serve as an emerging therapeutic target, and further research in this field will provide additional insights into the mechanisms and considerations for treatment of retinal pathologies.


Assuntos
Proteínas do Sistema Complemento , Doenças Retinianas , Humanos , Doenças Retinianas/tratamento farmacológico , Doenças Retinianas/fisiopatologia , Doenças Retinianas/imunologia , Proteínas do Sistema Complemento/fisiologia , Animais , Inativadores do Complemento/uso terapêutico , Inativadores do Complemento/farmacologia , Retinopatia Diabética/tratamento farmacológico , Retinopatia Diabética/fisiopatologia , Retina/efeitos dos fármacos , Retina/imunologia
4.
Stem Cell Reports ; 19(6): 817-829, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38729155

RESUMO

Several gaps and barriers remain for transplanting stem cells into the eye to treat ocular disease, especially diseases of the retina. While the eye has historically been considered immune privileged, recent thinking has identified the immune system as both a barrier and an opportunity for eye stem cell transplantation. Recent approaches leveraging scaffolds or cloaking have been considered in other tissues beyond immune suppression. This perspective paper outlines approaches for transplantation and proposes opportunities to overcome barriers of the immune system in stem cell transplantation in the eye.


Assuntos
Retina , Transplante de Células-Tronco , Humanos , Retina/imunologia , Retina/citologia , Transplante de Células-Tronco/métodos , Animais , Imunologia de Transplantes , Doenças Retinianas/terapia , Doenças Retinianas/imunologia
5.
Clin Interv Aging ; 19: 939-951, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38807637

RESUMO

Age-related macular degeneration (AMD) is a degenerative ocular disease primarily affecting central vision in the elderly. Its pathogenesis is complex, involving cellular senescence and immune homeostasis dysregulation. This review investigates the interaction between these two critical biological processes in AMD pathogenesis and their impact on disease progression. Initially, cellular senescence is analyzed, with particular emphasis on retinal damage induced by senescent retinal pigment epithelial cells. Subsequently, the occurrence of immune homeostasis dysregulation within the retina and its mechanistic role in AMD areis explored. Furthermore, the paper also discusses in detail the interplay between cellular senescence and immune responses, forming a vicious cycle that exacerbates retinal damage and may influence treatment outcomes. In summary, a deeper understanding of the interrelation between cellular senescence and immune dysregulation is vital for the developing innovative therapeutic strategies for AMD.


Assuntos
Senescência Celular , Homeostase , Degeneração Macular , Epitélio Pigmentado da Retina , Humanos , Degeneração Macular/imunologia , Epitélio Pigmentado da Retina/imunologia , Progressão da Doença , Retina/imunologia
6.
Immunol Lett ; 267: 106862, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38702033

RESUMO

BACKGROUND: Diabetic retinopathy (DR) stands as a prominent complication of diabetes. Berberine (BBR) has reported to be effective to ameliorate the retinal damage of DR. Studying the potential immunological mechanisms of BBR on the streptozotocin (STZ) induced DR mouse model will explain the therapeutic mechanisms of BBR and provide theoretical basis for the clinical application of this drug. METHODS: C57BL/6 J mice were induced into a diabetic state using a 50 mg/(kg·d) dose of STZ over a 5-day period. Subsequently, they were subjected to a high-fat diet (HFD) for one month. Following a 5-week treatment with 100 mg/(kg·d) BBR, the concentrations of inflammatory factors in the mice's peripheral blood were determined using an enzyme-linked immunosorbent assay (ELISA). Hematoxylin-eosin staining was employed to scrutinize pathological changes in the mice's retinas, while flow cytometry assessed the proportions of T-lymphocyte subsets and the activation status of dendritic cells (DCs) in the spleen and lymph nodes. CD4+T cells and DC2.4 cell lines were utilized to investigate the direct and indirect effects of BBR on T cells under high glucose conditions in vitro. RESULTS: Following 5 weeks of BBR treatment in the streptozotocin (STZ) mouse model of DR, we observed alleviation of retinal lesions and a down-regulation in the secretion of inflammatory cytokines, namely TNF-α, IL-1ß, and IL-6, in the serum of these mice. And in the spleen and lymph nodes of these mice, BBR inhibited the proportion of Th17 cells and promoted the proportion of Treg cells, thereby down-regulating the Th17/Treg ratio. Additionally, in vitro experiments, BBR directly inhibited the expression of the transcription factor RORγt and promoted the expression of the transcription factor Foxp3 in T cells, resulting in a down-regulation of the Th17/Treg ratio. Furthermore, BBR indirectly modulated the Th17/Treg ratio by suppressing the secretion of TNF-α, IL-1ß, and IL-6 by DCs and enhancing the secretion of indoleamine 2,3-dioxygenase (IDO) and transforming growth factor-beta (TGF-ß) by DCs. This dual action inhibited Th17 cell differentiation while promoting Treg cells. CONCLUSION: Our findings indicate that BBR regulate T cell subpopulation differentiation, reducing the Th17/Treg ratio by directly or indirectly pathway. This represents a potential therapeutic avenue of BBR for improving diabetic retinopathy.


Assuntos
Berberina , Diabetes Mellitus Experimental , Retinopatia Diabética , Linfócitos T Reguladores , Células Th17 , Animais , Berberina/farmacologia , Berberina/uso terapêutico , Retinopatia Diabética/tratamento farmacológico , Retinopatia Diabética/imunologia , Retinopatia Diabética/etiologia , Células Th17/imunologia , Células Th17/efeitos dos fármacos , Células Th17/metabolismo , Camundongos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/imunologia , Masculino , Citocinas/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Células Dendríticas/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Retina/patologia , Retina/imunologia , Retina/efeitos dos fármacos , Retina/metabolismo
7.
Int Immunopharmacol ; 134: 112231, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38739977

RESUMO

The roles of immune cell infiltration and ferroptosis in the progression of proliferative diabetic retinopathy (PDR) remain unclear. To identify upregulated molecules associated with immune infiltration and ferroptosis in PDR, GSE60436 and GSE102485 datasets were downloaded from the Gene Expression Omnibus (GEO). Genes associated with immune cell infiltration were examined through Weighted Gene Co-expression Network Analysis (WGCNA) and CIBERSORT algorithm. Common differentially expressed genes (DEGs) were intersected with ferroptosis-associated and immune cell infiltration-related genes. Localization of cellular expression was confirmed by single-cell analysis of GSE165784 dataset. Findings were validated by qRT-PCR, ELISA, Western blotting, and immunofluorescence staining. As a result, the infiltration of M2 macrophages was significantly elevated in fibrovascular membrane samples from PDR patients than the retinas of control subjects. Analysis of DEGs, M2 macrophage-related genes and ferroptosis-related genes identified three hub intersecting genes, TP53, HMOX1 and PPARA. qRT-PCR showed that HMOX1 was significantly higher in the oxygen-induced retinopathy (OIR) mouse model retinas than in controls. Single-cell analysis confirmed that HMOX1 was located in M2 macrophages. ELISA and western blotting revealed elevated levels of HMOX1 in the vitreous humor of PDR patients and OIR retinas, and immunofluorescence staining showed that HMOX1 co-localized with M2 macrophages in the retinas of OIR mice. This study offers novel insights into the mechanisms associated with immune cell infiltration and ferroptosis in PDR. HMOX1 expression correlated with M2 macrophage infiltration and ferroptosis, which may play a crucial role in PDR pathogenesis.


Assuntos
Retinopatia Diabética , Ferroptose , Heme Oxigenase-1 , Macrófagos , Regulação para Cima , Retinopatia Diabética/genética , Retinopatia Diabética/imunologia , Retinopatia Diabética/patologia , Retinopatia Diabética/metabolismo , Animais , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Humanos , Macrófagos/imunologia , Ferroptose/genética , Camundongos , Camundongos Endogâmicos C57BL , Retina/imunologia , Retina/patologia , Retina/metabolismo , Masculino , Modelos Animais de Doenças , Proteínas de Membrana
8.
Medicina (Kaunas) ; 60(5)2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38792993

RESUMO

Background and Objectives: Age-related macular degeneration (AMD) is one of the leading causes of central vision loss among elderly patients, and its dry form accounts for the majority of cases. Although several causes and mechanisms for the development and progression of AMD have previously been identified, the pathogenesis of this complex disease is still not entirely understood. As inflammation and immune system involvement are strongly suggested to play a central role in promoting the degenerative process and stimulating the onset of complications, we aimed to analyze the frequency of serum anti-retinal (ARAs) and anti-endothelial cell antibodies (AECAs) in patients with dry AMD and to determine their relationship with the clinical features of the disease, notably the area of geographic atrophy (GA). Materials and Methods: This study included 41 patients with advanced-stage dry AMD and 50 healthy controls without AMD, matched for gender and age. ARAs were detected by indirect immunofluorescence using monkey retina as an antigen substrate, and the presence of AECAs was determined using cultivated human umbilical vein endothelial cells and primate skeletal muscle. Results: ARAs were detected in 36 (87.8%) AMD patients (titers ranged from 1:20 to 1:320) and in 16 (39.0%) (titers ranged from 1:10 to 1:40) controls (p = 0.0000). Twenty of the forty-one patients (48.8%) were positive for AECAs, while in the control group, AECAs were present only in five sera (10.0%). The titers of AECAs in AMD patients ranged from 1:100 to 1:1000, and in the control group, the AECA titers were 1:100 (p = 0.0001). There were no significant correlations between the presence of AECAs and disease activity. Conclusions: This study demonstrates a higher prevalence of circulating AECAs in patients with dry AMD; however, no correlation was found between the serum levels of these autoantibodies and the area of GA.


Assuntos
Autoanticorpos , Atrofia Geográfica , Degeneração Macular , Humanos , Masculino , Feminino , Idoso , Atrofia Geográfica/sangue , Atrofia Geográfica/imunologia , Degeneração Macular/sangue , Degeneração Macular/complicações , Autoanticorpos/sangue , Pessoa de Meia-Idade , Idoso de 80 Anos ou mais , Células Endoteliais/imunologia , Retina/imunologia , Estudos de Casos e Controles
9.
Front Immunol ; 15: 1399989, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38799448

RESUMO

Introduction: Macrophage function is determined by microenvironment and origin. Brain and retinal microglia are both derived from yolk sac progenitors, yet their microenvironments differ. Utilizing single-cell RNA sequencing (scRNA-seq) data from mice, we tested the hypothesis that retinal and brain microglia exhibit distinct transcriptional profiles due to their unique microenvironments. Methods: Eyes and brains from 2-4 month wildtype mice were combined (20 eyes; 3 brains) to yield one biologically diverse sample per organ. Each tissue was digested into single cell suspensions, enriched for immune cells, and sorted for scRNA-seq. Analysis was performed in Seurat v3 including clustering, integration, and differential expression. Multi-parameter flow cytometry was used for validation of scRNA-seq results. Lymphocytic choriomeningitis virus (LCMV) Clone 13, which produces a systemic, chronic, and neurotropic infection, was used to validate scRNA-seq and flow cytometry results in vivo. Results: Cluster analysis of integrated gene expression data from eye and brain identified 6 Tmem119 + P2ry12 + microglial clusters. Differential expression analysis revealed that eye microglia were enriched for more pro-inflammatory processes including antigen processing via MHC class I (14.0-fold, H2-D1 and H2-K1) and positive regulation of T-cell immunity (8.4-fold) compared to brain microglia. Multi-parameter flow cytometry confirmed that retinal microglia expressed 3.2-fold greater H2-Db and 263.3-fold more H2-Kb than brain microglia. On Day 13 and 29 after LCMV infection, CD8+ T-cell density was greater in the retina than the brain. Discussion: Our data demonstrate that the microenvironment of retina and brain differs, resulting in microglia-specific gene expression changes. Specifically, retinal microglia express greater MHC class I by scRNA-seq and multi-parameter flow cytometry, resulting in a possibly enhanced capability to stimulate CD8+ T-cell inflammation during LCMV infection. These results may explain tissue-specific differences between retina and brain during systemic viral infections and CD8+ T-cell driven autoimmune disease.


Assuntos
Encéfalo , Microglia , Retina , Animais , Microglia/imunologia , Microglia/metabolismo , Camundongos , Retina/imunologia , Retina/patologia , Encéfalo/imunologia , Encéfalo/patologia , Encéfalo/metabolismo , Camundongos Endogâmicos C57BL , Vírus da Coriomeningite Linfocítica/imunologia , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Linfócitos T/imunologia , Inflamação/imunologia , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Análise de Célula Única , Linfócitos T CD8-Positivos/imunologia , Transcriptoma
10.
J Neuroinflammation ; 21(1): 112, 2024 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-38684986

RESUMO

BACKGROUND: Dimethyl fumarate (DMF) is a fumaric acid ester that exhibits immunoregulatory and anti-inflammatory properties. However, the function of DMF in autoimmune uveitis (AU) is incompletely understood, and studies comprehensively exploring the impact of DMF on immune cells are still lacking. METHODS: To explore the function of DMF in uveitis and its underlying mechanisms, we conducted single-cell RNA sequencing (scRNA-seq) on the cervical draining lymph node (CDLN) cells of normal, experimental autoimmune uveitis (EAU), and DMF-treated EAU mice. Additionally, we integrated scRNA-seq data of the retina and CDLNs to identify the potential impact of DMF on ocular immune cell infiltration. Flow cytometry was conducted to verify the potential target molecules of DMF. RESULTS: Our study showed that DMF treatment effectively ameliorated EAU symptoms. The proportional and transcriptional alterations in each immune cell type during EAU were reversed by DMF treatment. Bioinformatics analysis in our study indicated that the enhanced expression of Pim1 and Cxcr4 in EAU was reversed by DMF treatment. Further experiments demonstrated that DMF restored the balance between effector T (Teff) /regulatory T (Treg) cells through inhibiting the pathway of PIM1-protein kinase B (AKT)-Forkhead box O1 (FOXO1). By incorporating the scRNA-seq data of the retina from EAU mice into analysis, our study identified that T cells highly expressing Pim1 and Cxcr4 were enriched in the retina. DMF repressed the ocular infiltration of Teff cells, and this effect might depend on its inhibition of PIM1 and CXCR4 expression. Additionally, our study indicated that DMF might reduce the proportion of plasma cells by inhibiting PIM1 expression in B cells. CONCLUSIONS: DMF effectively attenuated EAU symptoms. During EAU, DMF reversed the Teff/Treg cell imbalance and suppressed the ocular infiltration of Teff cells by inhibiting PIM1 and CXCR4 expression. Thus, DMF may act as a new drug option for the treatment of AU.


Assuntos
Anti-Inflamatórios não Esteroides , Doenças Autoimunes , Fumarato de Dimetilo , Imunossupressores , Retina , Uveíte , Fumarato de Dimetilo/administração & dosagem , Fumarato de Dimetilo/farmacologia , Uveíte/genética , Uveíte/imunologia , Uveíte/terapia , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/terapia , Análise da Expressão Gênica de Célula Única , Modelos Animais de Doenças , Animais , Camundongos , Feminino , Camundongos Endogâmicos C57BL , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/genética , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Transcrição Gênica , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Atlas como Assunto , Imunossupressores/administração & dosagem , Imunossupressores/farmacologia , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/farmacologia , Retina/efeitos dos fármacos , Retina/imunologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia
11.
Int Immunopharmacol ; 133: 112021, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38626549

RESUMO

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.


Assuntos
Retinopatia Diabética , Quempferóis , Macrófagos , Microglia , Animais , Retinopatia Diabética/tratamento farmacológico , Retinopatia Diabética/imunologia , Retinopatia Diabética/patologia , Microglia/efeitos dos fármacos , Microglia/imunologia , Quempferóis/farmacologia , Quempferóis/uso terapêutico , Ratos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Progressão da Doença , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/imunologia , Retina/efeitos dos fármacos , Retina/patologia , Retina/imunologia , Linhagem Celular , Masculino , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Humanos , Agentes de Imunomodulação/farmacologia , Agentes de Imunomodulação/uso terapêutico , Modelos Animais de Doenças
12.
PLoS Pathog ; 20(4): e1012156, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38598560

RESUMO

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.


Assuntos
Barreira Hematorretiniana , COVID-19 , Retina , SARS-CoV-2 , SARS-CoV-2/imunologia , SARS-CoV-2/fisiologia , Animais , Barreira Hematorretiniana/virologia , COVID-19/imunologia , COVID-19/virologia , Camundongos , Humanos , Retina/virologia , Retina/imunologia , Retina/metabolismo , Enzima de Conversão de Angiotensina 2/metabolismo , Internalização do Vírus , Glicoproteína da Espícula de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/imunologia , Inflamação/imunologia , Inflamação/virologia , Betacoronavirus/fisiologia , Tropismo Viral , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Infecções por Coronavirus/patologia
13.
Front Immunol ; 15: 1374617, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38665911

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Vírus da Coriomeningite Linfocítica , Proteômica , Degeneração Retiniana , Animais , Camundongos , Proteômica/métodos , Degeneração Retiniana/imunologia , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos Endogâmicos C57BL , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Espectrometria de Massas em Tandem , Proteoma , Retina/imunologia , Retina/metabolismo , Retina/patologia , Cromatografia Líquida , Corioide/imunologia , Corioide/patologia , Corioide/metabolismo
14.
Acta Diabetol ; 61(7): 897-907, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38530415

RESUMO

OBJECTIVE: To examine the association between islet autoantibodies (IAbs) and the retinal neurovascular changes in type 1 diabetes mellitus (T1DM) with no diabetic retinopathy (NDR). METHODS: This cross-sectional study measured the neural retinal structure and microvascular density of 118 NDR eyes using spectral-domain optical coherence tomography angiography. Retinal structure parameters included retinal thickness (RT), inner retinal thickness (iRT), retina never fibral layer thickness (RNFL thickness), ganglion cell complex thickness (GCC thickness), and loss volume of GCC. Microvascular parameters included vessel density of superficial capillary plexus (sVD), vessel density of deep capillary plexus, and vessel density of choroid capillary plexus. Comparison and correlation analyses of these OCTA parameters were made with various IAbs, including glutamic acid decarboxylase antibody (GADA), tyrosine phosphatase-related islet antigen 2 antibody (IA2A), and zinc transporter 8 antibody (ZnT8A). A general linear model was used to understand the association of IAbs with the retina parameters. RESULTS: The IAb positive (IAbs +) group, which included 85 patients, had thinner RT (235.20 ± 18.10 mm vs. 244.40 ± 19.90 mm at fovea, P = 0.021) and thinner iRT (120.10 ± 9.00 mm vs. 124.70 ± 6.90 mm at parafovea, P = 0.015), compared with the IAb negative (IAbs-) group comprising 33 patients. Furthermore, a more severe reduction of RT was demonstrated in the presence of multiple IAbs. Among the three IAbs, GADA was the most significant independent risk factor of all-round RT decrease (ß = -0.20 vs. -0.27 at fovea and parafovea, respectively, P < 0.05), while titers of IA2A negatively affect sVD in the parafovea (ß = -0.316, P = 0.003). CONCLUSIONS: IAbs are associated with neural retinal thinning and microcirculation reduction in T1DM patients before the clinical onset of diabetic retinopathy.


Assuntos
Autoanticorpos , Diabetes Mellitus Tipo 1 , Retinopatia Diabética , Microcirculação , Retina , Humanos , Autoanticorpos/sangue , Autoanticorpos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/diagnóstico por imagem , Masculino , Feminino , Estudos Transversais , Adulto , Retinopatia Diabética/imunologia , Retinopatia Diabética/patologia , Retinopatia Diabética/diagnóstico por imagem , Retina/diagnóstico por imagem , Retina/imunologia , Retina/patologia , Pessoa de Meia-Idade , Tomografia de Coerência Óptica , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/diagnóstico por imagem , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/irrigação sanguínea , Vasos Retinianos/diagnóstico por imagem , Vasos Retinianos/patologia , Adulto Jovem
15.
J Mol Med (Berl) ; 102(5): 585-597, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38429382

RESUMO

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.


Assuntos
Retinopatia Diabética , Ativação de Macrófagos , Macrófagos , Retinopatia Diabética/imunologia , Retinopatia Diabética/patologia , Humanos , Animais , Macrófagos/imunologia , Macrófagos/metabolismo , Ativação de Macrófagos/imunologia , Retina/patologia , Retina/imunologia , Retina/metabolismo , Microglia/imunologia , Microglia/patologia , Microglia/metabolismo , Monócitos/imunologia , Monócitos/metabolismo
16.
Vis Neurosci ; 39: E005, 2022 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-36164752

RESUMO

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.


Assuntos
Interleucina-6 , Neuroglia , Lesões por Radiação , Retina , Degeneração Retiniana , Animais , Quimiocinas/genética , Quimiocinas/metabolismo , Interleucina-6/metabolismo , Luz , Neuroglia/imunologia , RNA Mensageiro/genética , Lesões por Radiação/etiologia , Lesões por Radiação/imunologia , Ratos , Retina/imunologia , Retina/efeitos da radiação , Degeneração Retiniana/etiologia , Degeneração Retiniana/imunologia
17.
Biochem Pharmacol ; 197: 114917, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35041813

RESUMO

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.


Assuntos
Doenças Autoimunes/prevenção & controle , Ganciclovir/uso terapêutico , Mediadores da Inflamação/antagonistas & inibidores , Retina/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Uveíte/prevenção & controle , Animais , Doenças Autoimunes/induzido quimicamente , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Progressão da Doença , Relação Dose-Resposta a Droga , Proteínas do Olho/toxicidade , Ganciclovir/farmacologia , Humanos , Mediadores da Inflamação/imunologia , Masculino , Ratos , Ratos Endogâmicos Lew , Retina/imunologia , Retina/patologia , Proteínas de Ligação ao Retinol/toxicidade , Células Th17/imunologia , Células Th17/patologia , Uveíte/induzido quimicamente , Uveíte/imunologia , Uveíte/patologia
18.
Invest Ophthalmol Vis Sci ; 62(15): 31, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34967854

RESUMO

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.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Sistema Imunitário/fisiologia , Interleucinas/uso terapêutico , Células Th17/imunologia , Uveíte/tratamento farmacológico , Transferência Adotiva , Animais , Células Apresentadoras de Antígenos/imunologia , Doenças Autoimunes/induzido quimicamente , Doenças Autoimunes/imunologia , Linfócitos B/imunologia , Técnicas de Cocultura , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Injeções Intravenosas , Interleucina-23/metabolismo , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Pescoço , Proteínas Recombinantes/uso terapêutico , Retina/imunologia , Análise de Sequência de RNA , Análise de Célula Única , Linfócitos T/imunologia , Células Th1/imunologia , Uveíte/induzido quimicamente , Uveíte/imunologia
19.
Cells ; 10(12)2021 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-34944083

RESUMO

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.


Assuntos
Ativação do Complemento/imunologia , Via Clássica do Complemento/imunologia , Hipóxia/imunologia , Retina/imunologia , Retina/patologia , Células Ganglionares da Retina/patologia , Animais , Apoptose/efeitos dos fármacos , Cobalto/toxicidade , Ativação do Complemento/efeitos dos fármacos , Via Alternativa do Complemento/efeitos dos fármacos , Via Alternativa do Complemento/imunologia , Via Clássica do Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/metabolismo , Peróxido de Hidrogênio/toxicidade , Lectinas/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Estresse Oxidativo/efeitos dos fármacos , Células Ganglionares da Retina/efeitos dos fármacos , Neurônios Retinianos/efeitos dos fármacos , Neurônios Retinianos/patologia , Estresse Fisiológico/efeitos dos fármacos , Suínos
20.
Front Immunol ; 12: 761776, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745138

RESUMO

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.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Microglia/imunologia , Animais , Receptor 1 de Quimiocina CX3C/genética , Encefalomielite Autoimune Experimental/diagnóstico por imagem , Feminino , Proteínas de Fluorescência Verde/genética , Masculino , Camundongos Transgênicos , Oftalmoscopia/métodos , Retina/diagnóstico por imagem , Retina/imunologia , Tomografia de Coerência Óptica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...