Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 133(8)2023 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-36862503

RESUMO

Defects in primary or motile cilia result in a variety of human pathologies, and retinal degeneration is frequently associated with these so-called ciliopathies. We found that homozygosity for a truncating variant in CEP162, a centrosome and microtubule-associated protein required for transition zone assembly during ciliogenesis and neuronal differentiation in the retina, caused late-onset retinitis pigmentosa in 2 unrelated families. The mutant CEP162-E646R*5 protein was expressed and properly localized to the mitotic spindle, but it was missing from the basal body in primary and photoreceptor cilia. This impaired recruitment of transition zone components to the basal body and corresponded to complete loss of CEP162 function at the ciliary compartment, reflected by delayed formation of dysmorphic cilia. In contrast, shRNA knockdown of Cep162 in the developing mouse retina increased cell death, which was rescued by expression of CEP162-E646R*5, indicating that the mutant retains its role for retinal neurogenesis. Human retinal degeneration thus resulted from specific loss of the ciliary function of CEP162.


Assuntos
Degeneração Retiniana , Animais , Humanos , Camundongos , Centrossomo/metabolismo , Cílios/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Neurogênese/genética , Retina/metabolismo , Degeneração Retiniana/metabolismo
2.
Methods Mol Biol ; 1834: 45-58, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30324435

RESUMO

Approximately 93 years ago at the zoological laboratories of Harvard University, Keeler, a medical geneticist, discovered a retina from a male albino mouse that was completely devoid of visual cells (rods). This rodless mouse was to be the first ever reported murine model of retinal degeneration. Over the years, naturally occurring retinal degeneration mouse mutants have been identified in several common laboratory inbred lines including FVB/NJ (Pde6brd1) and C57BL/6N (Crb1rd8). It is therefore imperative that vision researchers employing other genetically induced retinal degeneration models and experimental models such as laser-induced choroidal neovascularization (CNV) or bright white-light exposure screen for such naturally occurring mutations to prevent costly misinterpretations. In this regard, we describe herein simple molecular-based techniques for screening the presence of some commonly encountered rd mutations (Pde6brd1, Crb1rd8, Pde6brd10, and Rpe65rd12).


Assuntos
Estudos de Associação Genética , Predisposição Genética para Doença , Mutação , Degeneração Retiniana/diagnóstico , Degeneração Retiniana/genética , Alelos , Animais , Modelos Animais de Doenças , Estudos de Associação Genética/métodos , Loci Gênicos , Testes Genéticos , Genótipo , Camundongos , Camundongos Knockout , Fenótipo , Especificidade da Espécie
3.
Dis Model Mech ; 11(9)2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-30042155

RESUMO

Juvenile neuronal ceroid lipofuscinosis (jNCL) is a rare but fatal inherited lysosomal storage disorder mainly affecting children. The disease is caused by mutations in the CLN3 gene that lead to the accumulation of storage material in many tissues, prominent immune responses and neuronal degeneration. One of the first symptoms is vision loss followed by motor dysfunction and mental decline. The established Cln3Δex7/8 mouse model mimics many pathological features of the human disease except the retinal phenotype, which is very mild and occurs only very late in these mice. Here, we first carefully analyzed the retinal structure and microglia responses in these animals. While prominent autofluorescent spots were present in the fundus, only a moderate reduction of retinal thickness and no prominent microgliosis was seen in young CLN3-deficient mice. We next genetically introduced a light-sensitive RPE65 variant and established a light-damage paradigm that showed a high susceptibility of young Cln3Δex7/8 mice after exposure to 10,000 lux bright light for 30 min. Under these 'low light' conditions, CLN3-deficient mice showed a strong retinal degeneration, microglial activation, deposition of autofluorescent material and transcriptomic changes compared to wild-type animals. Finally, we treated the light-exposed Cln3Δex7/8 animals with the immunomodulatory compound minocycline, and thereby rescued the retinal phenotype and diminished microgliosis. Our findings indicate that exposure to specific light conditions accelerates CLN3-dependent retinal degeneration, and that immunomodulation by minocycline could be a possible treatment option to delay vision loss in jNCL patients.This article has an associated First Person interview with the first author of the paper.


Assuntos
Imunomodulação , Luz , Minociclina/uso terapêutico , Lipofuscinoses Ceroides Neuronais/tratamento farmacológico , Lipofuscinoses Ceroides Neuronais/imunologia , Degeneração Retiniana/tratamento farmacológico , Degeneração Retiniana/imunologia , Animais , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Fluorescência , Gliose/patologia , Inflamação/genética , Inflamação/patologia , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/patologia , Microglia/efeitos da radiação , Chaperonas Moleculares/metabolismo , Família Multigênica , Lipofuscinoses Ceroides Neuronais/genética , Lipofuscinoses Ceroides Neuronais/patologia , Fenótipo , Retina/metabolismo , Retina/patologia , Retina/efeitos da radiação , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Tomografia de Coerência Óptica , Transcriptoma/genética
4.
Sci Rep ; 8(1): 5970, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29654250

RESUMO

Mutations in the Norrin (NDP) gene cause severe developmental blood vessel defects in the retina leading to congenital blindness. In the retina of Ndph-knockout mice only the superficial capillary network develops. Here, a detailed characterization of this mouse model at late stages of the disease using in vivo retinal imaging revealed cystoid structures that closely resemble the ovoid cysts in the inner nuclear layer of the human retina with cystoid macular edema (CME). In human CME an involvement of Müller glia cells is hypothesized. In Ndph-knockout retinae we could demonstrate that activated Müller cells were located around and within these cystoid spaces. In addition, we observed extensive activation of retinal microglia and development of neovascularization. Furthermore, ex vivo analyses detected extravasation of monocytic cells suggesting a breakdown of the blood retina barrier. Thus, we could demonstrate that also in the developmental retinal vascular pathology present in the Ndph-knockout mouse inflammatory processes are active and may contribute to further retinal degeneration. This observation delivers a new perspective for curative treatments of retinal vasculopathies. Modulation of inflammatory responses might reduce the symptoms and improve visual acuity in these diseases.


Assuntos
Proteínas do Olho/metabolismo , Inflamação/patologia , Edema Macular/patologia , Neovascularização Patológica/patologia , Proteínas do Tecido Nervoso/metabolismo , Retina/patologia , Animais , Barreira Hematorretiniana/metabolismo , Barreira Hematorretiniana/patologia , Modelos Animais de Doenças , Humanos , Inflamação/metabolismo , Edema Macular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/metabolismo , Retina/metabolismo , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Acuidade Visual/fisiologia
5.
Methods Mol Biol ; 1753: 159-166, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29564787

RESUMO

Microgliosis is a hallmark of degenerative processes in the retina. Reactive microglia migrate to the photoreceptor layer and the subretinal space during outer retinal degeneration. This process creates a toxic milieu where reactive microglia and dying photoreceptors recruit additional reactive phagocytes. This results in the release of a multitude of proinflammatory factors which accelerate photoreceptor demise. In this chapter, we outline in detail how to monitor microgliosis in the Fam161a-deficient mouse model of Retinitis Pigmentosa by performing immunohistochemical stainings of retinal cryosections and flat mounts using the marker Iba1. This protocol will serve as a guideline in evaluating microglia reactivity and localization in various mouse models of retinal degeneration.


Assuntos
Imuno-Histoquímica/métodos , Microglia/patologia , Células Fotorreceptoras/patologia , Retinose Pigmentar/patologia , Animais , Proteínas de Ligação ao Cálcio/análise , Modelos Animais de Doenças , Proteínas do Olho/genética , Humanos , Imuno-Histoquímica/instrumentação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/análise , Retinose Pigmentar/genética
6.
Biochem Biophys Rep ; 12: 245-250, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29214225

RESUMO

Microglia activation plays an important role in immune responses in the CNS including the retina. Crocin, a plant-derived carotenoid, has been reported to possess anti-inflammatory, anti-apoptotic and anti-oxidative capacity in models of retinal damage and degeneration. If these neuroprotective effects could be mediated by direct modulation of microglial cells is unclear. Here, we examined the direct effects of crocin on key functions and pro-inflammatory gene expression in lipopolysaccharide (LPS)-activated BV-2 microglia. We found that crocin stimulation strongly promoted filopodia formation and markedly increased microglial phagocytosis, two important parameters relevant for physiological microglia functions. Moreover, crocin significantly reduced gene expression of the pro-inflammatory markers IL6, CCL2, and iNOS in LPS-challenged BV-2 cells and potently blocked NO production in these microglia. The observed immunomodulatory effects of crocin were not mediated by general inhibition of NFkB nuclear translocation. Our findings indicate that many of the anti-inflammatory effects of crocin demonstrated in animal models of neuronal degeneration could be mediated by its direct effects on microglia homeostasis.

7.
J Neuroinflammation ; 14(1): 4, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-28086806

RESUMO

BACKGROUND: Age-related macular degeneration (AMD) is the leading cause of blindness in developed countries. The polymorphism rs10490924 in the ARMS2 gene is highly associated with AMD and linked to an indel mutation (del443ins54), the latter inducing mRNA instability. At present, the function of the ARMS2 protein, the exact cellular sources in the retina and the biological consequences of the rs10490924 polymorphism are unclear. METHODS: Recombinant ARMS2 was expressed in Pichia pastoris, and protein functions were studied regarding cell surface binding and complement activation in human serum using fluoresence-activated cell sorting (FACS) as well as laser scanning microscopy (LSM). Biolayer interferometry defined protein interactions. Furthermore, endogenous ARMS2 gene expression was studied in human blood derived monocytes and in human induced pluripotent stem cell-derived microglia (iPSdM) by PCR and LSM. The ARMS2 protein was localized in human genotyped retinal sections and in purified monocytes derived from AMD patients without the ARMS2 risk variant by LSM. ARMS2 expression in monocytes under oxidative stress was determined by Western blot analysis. RESULTS: Here, we demonstrate for the first time that ARMS2 functions as surface complement regulator. Recombinant ARMS2 binds to human apoptotic and necrotic cells and initiates complement activation by recruiting the complement activator properdin. ARMS2-properdin complexes augment C3b surface opsonization for phagocytosis. We also demonstrate for the first time expression of ARMS2 in human monocytes especially under oxidative stress and in microglia cells of the human retina. The ARMS2 protein is absent in monocytes and also in microglia cells, derived from patients homozygous for the ARMS2 AMD risk variant (rs10490924). CONCLUSIONS: ARMS2 is likely involved in complement-mediated clearance of cellular debris. As AMD patients present with accumulated proteins and lipids on Bruch's membrane, ARMS2 protein deficiency due to the genetic risk variant might be involved in drusen formation.


Assuntos
Proteínas do Sistema Complemento/metabolismo , Degeneração Macular/genética , Degeneração Macular/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Proteínas/genética , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Animais , Células CHO , Proteínas do Sistema Complemento/genética , Cricetulus , Feminino , Heparitina Sulfato/farmacologia , Humanos , Peróxido de Hidrogênio/farmacologia , Fatores Imunológicos/farmacologia , Degeneração Macular/patologia , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Pessoa de Meia-Idade , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Properdina/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Proteínas/imunologia , Proteínas/metabolismo , Retina/metabolismo , Retina/patologia , Adulto Jovem
8.
Am J Hum Genet ; 99(2): 470-80, 2016 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-27486781

RESUMO

Inherited retinal dystrophies (iRDs) are a group of genetically and clinically heterogeneous conditions resulting from mutations in over 250 genes. Here, homozygosity mapping and whole-exome sequencing (WES) in a consanguineous family revealed a homozygous missense mutation, c.973C>T (p.His325Tyr), in RCBTB1. In affected individuals, it was found to segregate with retinitis pigmentosa (RP), goiter, primary ovarian insufficiency, and mild intellectual disability. Subsequent analysis of WES data in different cohorts uncovered four additional homozygous missense mutations in five unrelated families in whom iRD segregates with or without syndromic features. Ocular phenotypes ranged from typical RP starting in the second decade to chorioretinal dystrophy with a later age of onset. The five missense mutations affect highly conserved residues either in the sixth repeat of the RCC1 domain or in the BTB1 domain. A founder haplotype was identified for mutation c.919G>A (p.Val307Met), occurring in two families of Mediterranean origin. We showed ubiquitous mRNA expression of RCBTB1 and demonstrated predominant RCBTB1 localization in human inner retina. RCBTB1 was very recently shown to be involved in ubiquitination, more specifically as a CUL3 substrate adaptor. Therefore, the effect on different components of the CUL3 and NFE2L2 (NRF2) pathway was assessed in affected individuals' lymphocytes, revealing decreased mRNA expression of NFE2L2 and several NFE2L2 target genes. In conclusion, our study puts forward mutations in RCBTB1 as a cause of autosomal-recessive non-syndromic and syndromic iRD. Finally, our data support a role for impaired ubiquitination in the pathogenetic mechanism of RCBTB1 mutations.


Assuntos
Alelos , Fatores de Troca do Nucleotídeo Guanina/genética , Mutação de Sentido Incorreto/genética , Distrofias Retinianas/genética , Ubiquitinação/genética , Adolescente , Adulto , Idade de Início , Criança , Consanguinidade , Proteínas Culina/metabolismo , Exoma/genética , Feminino , Efeito Fundador , Genes Recessivos , Haplótipos/genética , Homozigoto , Humanos , Linfócitos/metabolismo , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , Linhagem , Fenótipo , RNA Mensageiro/genética , Retina/metabolismo , Síndrome , Turquia
9.
Sci Rep ; 6: 21307, 2016 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-26887858

RESUMO

The aim of this study was to unravel the molecular pathogenesis of an unusual retinitis pigmentosa (RP) phenotype observed in a Turkish consanguineous family. Homozygosity mapping revealed two candidate genes, SAMD7 and RHO. A homozygous RHO mutation c.448G > A, p.E150K was found in two affected siblings, while no coding SAMD7 mutations were identified. Interestingly, four non-coding homozygous variants were found in two SAMD7 genomic regions relevant for binding of the retinal transcription factor CRX (CRX-bound regions, CBRs) in these affected siblings. Three variants are located in a promoter CBR termed CBR1, while the fourth is located more downstream in CBR2. Transcriptional activity of these variants was assessed by luciferase assays and electroporation of mouse retinal explants with reporter constructs of wild-type and variant SAMD7 CBRs. The combined CBR2/CBR1 variant construct showed significantly decreased SAMD7 reporter activity compared to the wild-type sequence, suggesting a cis-regulatory effect on SAMD7 expression. As Samd7 is a recently identified Crx-regulated transcriptional repressor in retina, we hypothesize that these SAMD7 variants might contribute to the retinal phenotype observed here, characterized by unusual, recognizable pigment deposits, differing from the classic spicular intraretinal pigmentation observed in other individuals homozygous for p.E150K, and typically associated with RP in general.


Assuntos
Proteínas de Homeodomínio , Mutação de Sentido Incorreto , Elementos de Resposta , Retinose Pigmentar , Rodopsina , Transativadores , Substituição de Aminoácidos , Animais , Feminino , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Camundongos , Gravidez , Domínios Proteicos , Retinose Pigmentar/genética , Retinose Pigmentar/metabolismo , Rodopsina/genética , Rodopsina/metabolismo , Transativadores/genética , Transativadores/metabolismo , Turquia
10.
Biochem Biophys Res Commun ; 464(2): 434-40, 2015 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-26129774

RESUMO

Mutations in the acid sphingomyelinase (aSMase) coding gene sphingomyelin phosphodiesterase 1 (SMPD1) cause Niemann-Pick disease (NPD) type A and B. Sphingomyelin storage in cells of the mononuclear phagocyte system cause hepatosplenomegaly and severe neurodegeneration in the brain of NPD patients. However, the effects of aSMase deficiency on retinal structure and microglial behavior have not been addressed in detail yet. Here, we demonstrate that retinas of aSMase(-/-) mice did not display overt neuronal degeneration but showed significantly reduced scotopic and photopic responses in electroretinography. In vivo fundus imaging of aSMase(-/-) mice showed many hyperreflective spots and staining for the retinal microglia marker Iba1 revealed massive proliferation of retinal microglia that had significantly enlarged somata. Nile red staining detected prominent phospholipid inclusions in microglia and lipid analysis showed significantly increased sphingomyelin levels in retinas of aSMase(-/-) mice. In conclusion, the aSMase-deficient mouse is the first example in which microglial lipid inclusions are directly related to a loss of retinal function.


Assuntos
Microglia/enzimologia , Retina/enzimologia , Esfingomielina Fosfodiesterase/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Microglia/fisiologia , Fosfolipídeos/metabolismo , Retina/metabolismo , Retina/fisiopatologia , Esfingomielina Fosfodiesterase/genética
11.
Hum Mol Genet ; 23(19): 5197-210, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24833722

RESUMO

Mutations in the FAM161A gene were previously identified as the cause for autosomal-recessive retinitis pigmentosa 28. To study the effects of Fam161a dysfunction in vivo, we generated gene-trapped Fam161a(GT/GT) mice with a disruption of its C-terminal domain essential for protein-protein interactions. We confirmed the absence of the full-length Fam161a protein in the retina of Fam161a(GT/GT) mice using western blots and showed weak expression of a truncated Fam161a protein by immunohistochemistry. Histological analyses demonstrated that photoreceptor segments were disorganized in young Fam161a(GT/GT) mice and that the outer retina was completely lost at 6 months of age. Reactive microglia appeared in the outer retina and electroretinography showed an early loss of photoreceptor function in 4-month-old Fam161a(GT/GT) animals. Light and electron microscopy revealed a remarkable phenotype of a significantly shortened connecting cilium, spread ciliary microtubule doublets and disturbed disk organization in Fam161a(GT/GT) photoreceptor cells. Co-immunolabeling experiments demonstrated reduced expression and mislocalization of centrin 3 and disturbed targeting of the Fam161a interactors lebercilin and Cep290, which were restricted to the basal body and proximal connecting cilium in Fam161a(GT/GT) retinas. Moreover, we identified misrouting of the outer segment cargo proteins opsin and rds/peripherin 2 in Fam161a(GT/GT) mice. In conclusion, our results suggest a critical role for the C-terminal domain of Fam161a for molecular interactions and integrity of the connecting cilium. Fam161a is required for the molecular delivery into the outer segment cilium, a function which is essential for outer segment disk formation and ultimately visual function.


Assuntos
Proteínas do Olho/genética , Mutação , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patologia , Degeneração Retiniana/genética , Potenciais de Ação , Animais , Proteínas de Transporte/metabolismo , Feminino , Expressão Gênica , Marcação de Genes , Loci Gênicos , Genótipo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Células Fotorreceptoras/ultraestrutura , Ligação Proteica , Transporte Proteico , Retina/metabolismo , Degeneração Retiniana/patologia , Degeneração Retiniana/fisiopatologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Transtornos da Visão/genética , Transtornos da Visão/patologia , Transtornos da Visão/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...