Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Development ; 151(11)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38682276

RESUMEN

The GPR124/RECK/WNT7 pathway is an essential regulator of CNS angiogenesis and blood-brain barrier (BBB) function. GPR124, a brain endothelial adhesion seven-pass transmembrane protein, associates with RECK, which binds and stabilizes newly synthesized WNT7 that is transferred to frizzled (FZD) to initiate canonical ß-catenin signaling. GPR124 remains enigmatic: although its extracellular domain (ECD) is essential, the poorly conserved intracellular domain (ICD) appears to be variably required in mammals versus zebrafish, potentially via adaptor protein bridging of GPR124 and FZD ICDs. GPR124 ICD deletion impairs zebrafish angiogenesis, but paradoxically retains WNT7 signaling upon mammalian transfection. We thus investigated GPR124 ICD function using the mouse deletion mutant Gpr124ΔC. Despite inefficiently expressed GPR124ΔC protein, Gpr124ΔC/ΔC mice could be born with normal cerebral cortex angiogenesis, in comparison with Gpr124-/- embryonic lethality, forebrain avascularity and hemorrhage. Gpr124ΔC/ΔC vascular phenotypes were restricted to sporadic ganglionic eminence angiogenic defects, attributable to impaired GPR124ΔC protein expression. Furthermore, Gpr124ΔC and the recombinant GPR124 ECD rescued WNT7 signaling in culture upon brain endothelial Gpr124 knockdown. Thus, in mice, GPR124-regulated CNS forebrain angiogenesis and BBB function are exerted by ICD-independent functionality, extending the signaling mechanisms used by adhesion seven-pass transmembrane receptors.


Asunto(s)
Barrera Hematoencefálica , Encéfalo , Neovascularización Fisiológica , Receptores Acoplados a Proteínas G , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/embriología , Neovascularización Fisiológica/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Ratones , Encéfalo/metabolismo , Encéfalo/embriología , Dominios Proteicos , Ratones Noqueados , Transducción de Señal , Proteínas Wnt/metabolismo , Proteínas Wnt/genética , Humanos , Células Endoteliales/metabolismo , Angiogénesis , Proteínas Ligadas a GPI
2.
Klin Monbl Augenheilkd ; 240(5): 650-661, 2023 May.
Artículo en Inglés, Alemán | MEDLINE | ID: mdl-37207638

RESUMEN

Complex barriers comprise the blood-aqueous (BAB) and the blood-retinal barrier (BRB), and separate anterior and posterior eye chambers, vitreous body, and sensory retina from the circulation. They prevent pathogens and toxins from entering the eye, control movement of fluid, proteins, and metabolites, and contribute to the maintenance of the ocular immune status. Morphological correlates of blood-ocular barriers are tight junctions between neighboring endothelial and epithelial cells, which function as gatekeepers of the paracellular transport of molecules, thereby limiting their uncontrolled access to ocular chambers and tissues. The BAB is composed of tight junctions between endothelial cells of the iris vasculature, endothelial cells of Schlemm's canal inner wall, and cells of the nonpigmented ciliary epithelium. The BRB consists of tight junctions between endothelial cells of the retinal vessels (inner BRB) and epithelial cells of the retinal pigment epithelium (outer BRB). These junctional complexes respond rapidly to pathophysiological changes, thus enabling vascular leakage of blood-derived molecules and inflammatory cells into ocular tissues and chambers. Blood-ocular barrier function, which can be clinically measured by laser flare photometry or fluorophotometry, is compromised in traumatic, inflammatory, or infectious processes, but also frequently contributes to the pathophysiology of chronic diseases of the anterior eye segment and the retina, as exemplified by diabetic retinopathy and age-related macular degeneration.


Asunto(s)
Retinopatía Diabética , Células Endoteliales , Humanos , Retina , Barrera Hematorretinal/fisiología , Epitelio Pigmentado de la Retina
3.
Int J Mol Sci ; 23(7)2022 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-35408803

RESUMEN

Macular neovascularization type 3, formerly known as retinal angiomatous proliferation (RAP), is a hallmark of age-related macular degeneration and is associated with an accumulation of myeloid cells, such as microglia (MG) and infiltrating blood-derived macrophages (MAC). However, the contribution of MG and MAC to the myeloid cell pool at RAP sites and their exact functions remain unknown. In this study, we combined a microglia-specific reporter mouse line with a mouse model for RAP to identify the contribution of MG and MAC to myeloid cell accumulation at RAP and determined the transcriptional profile of MG using RNA sequencing. We found that MG are the most abundant myeloid cell population around RAP, whereas MAC are rarely, if ever, associated with late stages of RAP. RNA sequencing of RAP-associated MG showed that differentially expressed genes mainly contribute to immune-associated processes, including chemotaxis and migration in early RAP and proliferative capacity in late RAP, which was confirmed by immunohistochemistry. Interestingly, MG upregulated only a few angiomodulatory factors, suggesting a rather low angiogenic potential. In summary, we showed that MG are the dominant myeloid cell population at RAP sites. Moreover, MG significantly altered their transcriptional profile during RAP formation, activating immune-associated processes and exhibiting enhanced proliferation, however, without showing substantial upregulation of angiomodulatory factors.


Asunto(s)
Degeneración Macular , Neovascularización Retiniana , Animales , Proliferación Celular/genética , Angiografía con Fluoresceína , Degeneración Macular/complicaciones , Ratones , Microglía , Neovascularización Patológica/complicaciones , Neovascularización Retiniana/genética , Tomografía de Coherencia Óptica
4.
Int J Mol Sci ; 23(5)2022 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-35269767

RESUMEN

Transforming growth factor ß (TGFß) signaling has manifold functions such as regulation of cell growth, differentiation, migration, and apoptosis. Moreover, there is increasing evidence that it also acts in a neuroprotective manner. We recently showed that TGFß receptor type 2 (Tgfbr2) is upregulated in retinal neurons and Müller cells during retinal degeneration. In this study we investigated if this upregulation of TGFß signaling would have functional consequences in protecting retinal neurons. To this end, we analyzed the impact of TGFß signaling on photoreceptor viability using mice with cell type-specific deletion of Tgfbr2 in retinal neurons and Müller cells (Tgfbr2ΔOC) in combination with a genetic model of photoreceptor degeneration (VPP). We examined retinal morphology and the degree of photoreceptor degeneration, as well as alterations of the retinal transcriptome. In summary, retinal morphology was not altered due to TGFß signaling deficiency. In contrast, VPP-induced photoreceptor degeneration was drastically exacerbated in double mutant mice (Tgfbr2ΔOC; VPP) by induction of pro-apoptotic genes and dysregulation of the MAP kinase pathway. Therefore, TGFß signaling in retinal neurons and Müller cells exhibits a neuroprotective effect and might pose promising therapeutic options to attenuate photoreceptor degeneration in humans.


Asunto(s)
Degeneración Retiniana , Factor de Crecimiento Transformador beta , Animales , Modelos Animales de Enfermedad , Células Ependimogliales/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Retina/metabolismo , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
5.
Int J Mol Sci ; 23(15)2022 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-35897674

RESUMEN

Protocadherins (PCDHs) belong to the cadherin superfamily and represent the largest subgroup of calcium-dependent adhesion molecules. In the genome, most PCDHs are arranged in three clusters, α, ß, and γ on chromosome 5q31. PCDHs are highly expressed in the central nervous system (CNS). Several PCDHs have tumor suppressor functions, but their individual role in primary brain tumors has not yet been elucidated. Here, we examined the mRNA expression of PCDHGC3, a member of the PCDHγ cluster, in non-cancerous brain tissue and in gliomas of different World Health Organization (WHO) grades and correlated it with the clinical data of the patients. We generated a PCDHGC3 knockout U343 cell line and examined its growth rate and migration in a wound healing assay. We showed that PCDHGC3 mRNA and protein were significantly overexpressed in glioma tissue compared to a non-cancerous brain specimen. This could be confirmed in glioma cell lines. High PCDHGC3 mRNA expression correlated with longer progression-free survival (PFS) in glioma patients. PCDHGC3 knockout in U343 resulted in a slower growth rate but a significantly faster migration rate in the wound healing assay and decreased the expression of several genes involved in WNT signaling. PCDHGC3 expression should therefore be further investigated as a PFS-marker in gliomas. However, more studies are needed to elucidate the molecular mechanisms underlying the PCDHGC3 effects.


Asunto(s)
Neoplasias Encefálicas , Proteínas Relacionadas con las Cadherinas , Glioblastoma , Glioma , Neoplasias Encefálicas/genética , Proteínas Relacionadas con las Cadherinas/genética , Cadherinas/genética , Cadherinas/metabolismo , Glioblastoma/genética , Glioma/genética , Humanos , Supervivencia sin Progresión , Protocadherinas , ARN Mensajero
6.
Arterioscler Thromb Vasc Biol ; 40(1): 159-174, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31619060

RESUMEN

OBJECTIVE: In proliferative retinopathies, complications derived from neovascularization cause blindness. During early disease, pericyte's apoptosis contributes to endothelial dysfunction and leakage. Hypoxia then drives VEGF (vascular endothelial growth factor) secretion and pathological neoangiogenesis. Cardiac ANP (atrial natriuretic peptide) contributes to systemic microcirculatory homeostasis. ANP is also formed in the retina, with unclear functions. Here, we characterized whether endogenously formed ANP regulates retinal (neo)angiogenesis. Approach and Results: Retinal vascular development and ischemia-driven neovascularization were studied in mice with global deletion of GC-A (guanylyl cyclase-A), the cGMP (cyclic guanosine monophosphate)-forming ANP receptor. Mice with a floxed GC-A gene were interbred with Tie2-Cre, GFAP-Cre, or PDGF-Rß-CreERT2 lines to dissect the endothelial, astrocyte versus pericyte-mediated actions of ANP in vivo. In neonates with global GC-A deletion (KO), vascular development was mildly delayed. Moreover, such KO mice showed augmented vascular regression and exacerbated ischemia-driven neovascularization in the model of oxygen-induced retinopathy. Notably, absence of GC-A in endothelial cells did not impact retinal vascular development or pathological neovascularization. In vitro ANP/GC-A/cGMP signaling, via activation of cGMP-dependent protein kinase I, inhibited hypoxia-driven astrocyte's VEGF secretion and TGF-ß (transforming growth factor beta)-induced pericyte apoptosis. In neonates lacking ANP/GC-A signaling in astrocytes, vascular development and hyperoxia-driven vascular regression were unaltered; ischemia-induced neovascularization was modestly increased. Remarkably, inactivation of GC-A in pericytes retarded physiological retinal vascularization and markedly enhanced cell apoptosis, vascular regression, and subsequent neovascularization in oxygen-induced retinopathy. CONCLUSIONS: Protective pericyte effects of the ANP/GC-A/cGMP pathway counterregulate the initiation and progression of experimental proliferative retinopathy. Our observations indicate augmentation of endogenous pericyte ANP signaling as target for treatment of retinopathies associated with neovascularization.


Asunto(s)
Astrocitos/metabolismo , GMP Cíclico/genética , Regulación del Desarrollo de la Expresión Génica , Péptidos Natriuréticos/metabolismo , Pericitos/metabolismo , ARN/genética , Neovascularización Retiniana/genética , Animales , Animales Recién Nacidos , Apoptosis , Astrocitos/patología , Células Cultivadas , GMP Cíclico/biosíntesis , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Immunoblotting , Ratones , Ratones Transgénicos , Pericitos/patología , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Transducción de Señal
7.
Int J Mol Sci ; 22(12)2021 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-34208383

RESUMEN

Hereditary retinal degenerations like retinitis pigmentosa (RP) are among the leading causes of blindness in younger patients. To enable in vivo investigation of cellular and molecular mechanisms responsible for photoreceptor cell death and to allow testing of therapeutic strategies that could prevent retinal degeneration, animal models have been created. In this study, we deeply characterized the transcriptional profile of mice carrying the transgene rhodopsin V20G/P23H/P27L (VPP), which is a model for autosomal dominant RP. We examined the degree of photoreceptor degeneration and studied the impact of the VPP transgene-induced retinal degeneration on the transcriptome level of the retina using next generation RNA sequencing (RNASeq) analyses followed by weighted correlation network analysis (WGCNA). We furthermore identified cellular subpopulations responsible for some of the observed dysregulations using in situ hybridizations, immunofluorescence staining, and 3D reconstruction. Using RNASeq analysis, we identified 9256 dysregulated genes and six significantly associated gene modules in the subsequently performed WGCNA. Gene ontology enrichment showed, among others, dysregulation of genes involved in TGF-ß regulated extracellular matrix organization, the (ocular) immune system/response, and cellular homeostasis. Moreover, heatmaps confirmed clustering of significantly dysregulated genes coding for components of the TGF-ß, G-protein activated, and VEGF signaling pathway. 3D reconstructions of immunostained/in situ hybridized sections revealed retinal neurons and Müller cells as the major cellular population expressing representative components of these signaling pathways. The predominant effect of VPP-induced photoreceptor degeneration pointed towards induction of neuroinflammation and the upregulation of neuroprotective pathways like TGF-ß, G-protein activated, and VEGF signaling. Thus, modulation of these processes and signaling pathways might represent new therapeutic options to delay the degeneration of photoreceptors in diseases like RP.


Asunto(s)
Perfilación de la Expresión Génica , Neuroprotección/genética , Retinitis Pigmentosa/genética , Transcripción Genética , Regulación hacia Arriba/genética , Animales , Quimiocina CCL2/metabolismo , Femenino , Proteínas de Unión al GTP/metabolismo , Redes Reguladoras de Genes , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Ratones , Ratones Transgénicos , Neuroglía/metabolismo , Degeneración Retiniana/complicaciones , Degeneración Retiniana/patología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Rodopsina/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Exp Eye Res ; 186: 107710, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31254512

RESUMEN

Quantifying the number of axons in the optic nerve is of interest in many research questions. Here, we show that a stereological method allows simple, efficient, precise and unbiased determination of the total axon number in the murine optic nerve. Axons in semi-thin optic nerve cross sections from untreated eyes (n = 21) and eyes subjected to retinal damage by intravitreous NMDA injections (n = 32) or PBS controls (n = 5) were manually identified, counted and digitally labeled by hand. A stereological procedure was empirically tested with systematic combinations of different sampling methods (simple random sampling without replacement, systematic uniform random sampling, stratified random sampling) and sampling parameters. Extensive numerical Monte Carlo experiments were performed to evaluate their large-sample properties. Our results demonstrate reliable determination of total axon number and superior performance compared to other methods at a small fraction of the time required for a full manual count. We specify suitable sampling parameters for the adoption of an efficient stereological sampling scheme, give empirical estimates of the additionally introduced sampling variance to facilitate experimental planning, and offer AxonCounter, an easy-to-use plugin implementing these stereological methods for the multi-platform image processing application NIH ImageJ.


Asunto(s)
Recuento de Células/métodos , Técnicas Citológicas , Nervio Óptico/citología , Animales , Agonistas de Aminoácidos Excitadores/farmacología , Procesamiento de Imagen Asistido por Computador/métodos , Ratones , Ratones Endogámicos BALB C , N-Metilaspartato/farmacología , Nervio Óptico/efectos de los fármacos
9.
Exp Eye Res ; 179: 115-124, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30399364

RESUMEN

Sequence variants in LOXL1 coding for the secreted enzyme lysyl oxidase homolog 1 (LOXL1) associate with pseudoexfoliation (PEX) syndrome, a condition that is characterized by the deposition of extracellular fibrillar PEX material in the anterior eye and other parts of the body. Since the specific role of LOXL1 in the pathogenesis of PEX is unclear, and an increase in its expression was reported for early stages of PEX syndrome, we generated and studied transgenic mice with ocular overexpression of its mouse ortholog Loxl1. The chicken ßB1-crystallin promoter was used to overexpress Loxl1 in the lenses of ßB1-crystallin-Loxl1 transgenic mice. Transgenic lenses contained high levels of the protein LOXL1 and its mRNA, which were both not detectable in lenses of wildtype littermates. In wildtype mice, immunoreactivity for LOXL1 was mainly seen extracellularly in region of the ciliary zonules. ßB1-crystallin-Loxl1 littermates showed an additional diffuse immunostaining in lens fibers and capsule, and in the inner limiting membrane and retina indicating secretion of soluble LOXL1 from transgenic lenses. In addition, lens fibers of transgenic animals contained multiple distinct spots of very intense LOXL1 immunoreactivity. By transmission electron microscopy, those spots correlated with electron-dense round or oval bodies of 20-50 nm in diameter which were localized in the rough endoplasmic reticulum and not seen in wildtype lenses. Immunogold electron microscopy confirmed that the electron-dense bodies contained LOXL1 indicating aggregation of insoluble LOXL1. Similar structures were seen in the extracellular lens capsule suggesting their secretion from lens fibers. Otherwise, no changes were seen between the eyes of ßB1-crystallin-Loxl1 mice and their wildtype littermates, neither by light microscopy and funduscopy of whole eyes, nor by scanning and quantitative transmission electron microscopy of ciliary epithelium and zonules. At one month of age, intraocular pressure was significantly higher in transgenic mice than in wildtype littermates. No differences in IOP were seen though at 2-5 months of age. We conclude that LOXL1 has a strong tendency to aggregate in the rER when expressed in vivo at high amounts. A similar scenario, involving intracellular aggregation of LOXL1 and secretion of LOXL1 aggregates into the extracellular space, may be involved in the early pathogenetic events in eyes of PEX patients.


Asunto(s)
Aminoácido Oxidorreductasas/genética , Cuerpo Ciliar/metabolismo , Síndrome de Exfoliación/metabolismo , Regulación de la Expresión Génica/fisiología , Cristalino/metabolismo , Agregado de Proteínas/fisiología , Aminoácido Oxidorreductasas/metabolismo , Animales , Western Blotting , Cuerpo Ciliar/ultraestructura , Síndrome de Exfoliación/etiología , Femenino , Inmunohistoquímica , Presión Intraocular , Cápsula del Cristalino/metabolismo , Cristalino/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Cadena B de beta-Cristalina/genética
10.
Adv Exp Med Biol ; 1185: 295-299, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31884627

RESUMEN

The degeneration of photoreceptors is a common hallmark of ocular diseases like retinitis pigmentosa (RP) or age-related macular degeneration (AMD). To experimentally induce photoreceptor degeneration, the light damage paradigm is frequently used. In this study we show that the exposure to high amounts of cool white light (10,000 lux, 1 h) resulted in a more than 11-fold higher apoptotic rate in the retina compared to light exposure with 5000 lux for 30 min. Consequently, exposure to intense light resulted in a significant downregulation of retinal mRNA expression levels of the reference genes Gapdh, Gnb2l, Rpl32, Rps9, Actb, Ubc or Tbp compared to untreated controls. Investigators performing light-induced photoreceptor degeneration should be aware of the fact that higher light intensities will result in a dysregulation of reference genes.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Luz , Células Fotorreceptoras de Vertebrados/efectos de la radiación , Retina/efectos de la radiación , Apoptosis , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Células Fotorreceptoras de Vertebrados/patología , Retina/citología , Degeneración Retiniana/patología , Retinitis Pigmentosa/patología
11.
Adv Exp Med Biol ; 1185: 519-523, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31884664

RESUMEN

The vasoactive peptide endothelin is an effective regulator of blood pressure and vascular homeostasis. In addition, the dysregulation of the endothelin signaling pathway is discussed to contribute to ocular diseases like glaucoma or diabetic retinopathy. Furthermore, our workgroup and others showed a protective effect of endothelin 2 for the survival of photoreceptors. In this study, we analyzed mRNA expression levels of the endothelin signaling family in wild-type mice after a puncture of the eye, intravitreal PBS injections, or light-induced photoreceptor degeneration. We observed elevated endothelin receptor a (Eta), endothelin receptor b (Etb), endothelin 1(Et1), and endothelin 2 (Et2) levels, while endothelin 3 (Et3) mRNA levels were not significantly altered. Our findings indicate an important role of the endothelin signaling pathway in response to ocular trauma or disease. These findings make endothelin signaling a promising target to attenuate retinal degeneration.


Asunto(s)
Endotelinas/metabolismo , Oftalmopatías/patología , Receptores de Endotelina/metabolismo , Retina/metabolismo , Transducción de Señal , Animales , Ratones , Retina/patología
12.
Am J Pathol ; 187(11): 2570-2589, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28823871

RESUMEN

The molecular pathogenesis of choroidal neovascularization (CNV), an angiogenic process that critically contributes to vision loss in age-related macular degeneration, is unclear. Herein, we analyzed the role of transforming growth factor (TGF)-ß signaling for CNV formation by generating a series of mutant mouse models with induced conditional deletion of TGF-ß signaling in the entire eye, the retinal pigment epithelium (RPE), or the vascular endothelium. Deletion of TGF-ß signaling in the eye caused CNV, irrespectively if it was ablated in newborn or 3-week-old mice. Areas of CNV showed photoreceptor degeneration, multilayered RPE, basal lamina deposits, and accumulations of monocytes/macrophages. The changes progressed, leading to marked structural and functional alterations of the retina. Although the specific deletion of TGF-ß signaling in the RPE caused no obvious changes, specific deletion in vascular endothelial cells caused CNV and a phenotype similar to that observed after the deletion in the entire eye. We conclude that impairment of TGF-ß signaling in the vascular endothelium of the eye is sufficient to trigger CNV formation. Our findings highlight the importance of TGF-ß signaling as a key player in the development of ocular neovascularization and indicate a fundamental role of TGF-ß signaling in the pathogenesis of age-related macular degeneration.


Asunto(s)
Neovascularización Coroidal/metabolismo , Degeneración Macular/patología , Epitelio Pigmentado de la Retina/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Coroides/patología , Neovascularización Coroidal/genética , Modelos Animales de Enfermedad , Ratones Noqueados , Retina/metabolismo , Factor de Crecimiento Transformador beta/genética
13.
Histochem Cell Biol ; 147(4): 453-469, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27787612

RESUMEN

Olfactomedin 1 (OLFM1) is a secreted glycoprotein and member of the olfactomedin protein family, which is preferentially expressed in various areas throughout the central nervous system. To learn about the functional properties of OLFM1 in the eye, we investigated its localization in the mouse and pig eye. In addition, we analyzed the ocular phenotype of Olfm1 mutant mice in which 52 amino acids were deleted in the central part (M2 region) of OLFM1. OLFM1 was detected in cornea, sclera, retina, and optic nerve of both wild-type and Olfm1 mutant littermates. By immunohistochemistry and double labeling with the lectin peanut agglutinin, OLFM1 was found in the interphotoreceptor matrix (IPM) of mouse and pig retina where it was directly localized to the inner segments of photoreceptors. Western blotting confirmed the presence of the OLFM1 isoforms pancortin 1 (BMY) and pancortin 2 (BMZ) in the IPM. The retinal phenotype of Olfm1 mutant mice did not obviously differ from that of wild-type littermates. In addition, outer nuclear layer (ONL) and total retinal thickness were not different, and the same was true for the area of the optic nerve in cross sections. Functional changes were observed though by electroretinography, which showed significantly lower a- and b-wave amplitudes in Olfm1 mutant mice when compared to age-matched wild-type mice. When light damage experiments were performed as an experimental paradigm of photoreceptor apoptosis, significantly more TUNEL-positive cells were observed in Olfm1 mutant mice 30 h after light exposure. One week after light exposure, the ONL was significantly thinner in Olfm1 mutant mice than in wild-type littermates indicating increased photoreceptor loss. No differences were observed when rhodopsin turnover or ERK1/2 signaling was investigated. We conclude that OLFM1 is a newly identified IPM molecule that serves an important role for photoreceptor homeostasis, which is significantly compromised in the eyes of Olfm1 mutant mice.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de la radiación , Glicoproteínas/genética , Glicoproteínas/metabolismo , Luz/efectos adversos , Retina/patología , Retina/efectos de la radiación , Animales , Matriz Extracelular/patología , Ratones , Mutación , Células Fotorreceptoras/metabolismo , Retina/metabolismo
14.
Histochem Cell Biol ; 148(1): 21-32, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28258388

RESUMEN

The transforming growth factor-ß (TGF-ß) pathway contributes to maintain the quiescence of adult neural stem and progenitor cells in the brain. In the retina, Müller cells are discussed to represent a glial cell population with progenitor-like characteristics. Here, we aimed to investigate if elevated TGF-ß signaling modulates the proliferation of Müller cells during retinal development. We generated mutant mice with a systemic, heterozygous up-regulation of TGF-ß signaling by deleting its inhibitor SMAD7. We investigated apoptosis, proliferation, and differentiation of Müller cells in the developing retina. We show that a heterozygous deletion of SMAD7 results in an increased proliferation of Müller cell progenitors in the central retina at postnatal day 4, the time window when Müller cells differentiate in the mouse retina. This in turn results in a thickened retina and inner nuclear layer and a higher number of differentiated Müller cells in the more developed retina. Müller cells in mutant mice contain higher amounts of nestin than those of control animals which indicates that the increase in TGF-ß signaling activity during retinal development contribute to maintain some progenitor-like characteristics in Müller cells even after their differentiation period. We conclude that TGF-ß signaling influences Müller cell proliferation and differentiation during retinal development.


Asunto(s)
Proliferación Celular , Retina/crecimiento & desarrollo , Retina/metabolismo , Proteína smad7/deficiencia , Células Madre/citología , Células Madre/metabolismo , Animales , Diferenciación Celular , Ratones , Ratones Noqueados , Retina/citología , Factor de Crecimiento Transformador beta/metabolismo
15.
Am J Pathol ; 185(6): 1749-68, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25857227

RESUMEN

Diabetic retinopathy, a major cause of blindness, is characterized by a distinct phenotype. The molecular causes of the phenotype are not sufficiently clear. Here, we report that deletion of transforming growth factor ß signaling in the retinal microenvironment of newborn mice induces changes that largely mimic the phenotype of nonproliferative and proliferative diabetic retinopathy in humans. Lack of transforming growth factor ß signaling leads to the formation of abundant microaneurysms, leaky capillaries, and retinal hemorrhages. Retinal capillaries are not covered by differentiated pericytes, but by a coat of vascular smooth muscle-like cells and a thickened basal lamina. Reactive microglia is found in close association with retinal capillaries. In older animals, loss of endothelial cells and the formation of ghost vessels are observed, findings that correlate with the induction of angiogenic molecules and the accumulation of retinal hypoxia-inducible factor 1α, indicating hypoxia. Consequently, retinal and vitreal neovascularization occurs, a scenario that leads to retinal detachment, vitreal hemorrhages, neuronal apoptosis, and impairment of sensory function. We conclude that transforming growth factor ß signaling is required for the differentiation of retinal pericytes during vascular development of the retina. Lack of differentiated pericytes initiates a scenario of structural and functional changes in the retina that mimics those of diabetic retinopathy strongly indicating a common mechanism.


Asunto(s)
Apoptosis/fisiología , Retinopatía Diabética/metabolismo , Neovascularización Retiniana/metabolismo , Vasos Retinianos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Diferenciación Celular , Retinopatía Diabética/genética , Retinopatía Diabética/patología , Ratones , Ratones Transgénicos , Pericitos/metabolismo , Pericitos/patología , Neovascularización Retiniana/genética , Neovascularización Retiniana/patología , Vasos Retinianos/patología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/genética
16.
Adv Exp Med Biol ; 854: 495-500, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26427451

RESUMEN

Embryonic lethality in mice with targeted gene deletion is a major issue that can be circumvented by using Cre-loxP-based animal models. Various inducible Cre systems are available, e.g. such that are activated following tamoxifen treatment, and allow deletion of a specific target gene at any desired time point during the life span of the animal. In this study, we describe the efficiency of topical tamoxifen administration by eye drops using a Cre- reporter mouse strain (R26R). We report that tamoxifen-responsive CAGGCre-ER (TM) mice show a robust Cre- mediated recombination throughout the entire eye.


Asunto(s)
Ojo/efectos de los fármacos , Integrasas/metabolismo , Recombinación Genética/efectos de los fármacos , Tamoxifeno/farmacología , Animales , Antagonistas de Estrógenos/farmacología , Ojo/metabolismo , Integrasas/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Soluciones Oftálmicas/farmacología , beta-Galactosidasa/metabolismo
17.
J Cell Mol Med ; 19(5): 1010-20, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25704370

RESUMEN

Connective tissue growth factor (CTGF) induces extracellular matrix (ECM) synthesis and contractility in human trabecular meshwork (HTM) cells. Both processes are involved in the pathogenesis of primary open-angle glaucoma. To date, little is known about regulation and function of CTGF expression in the trabecular meshwork (TM). Therefore, we analysed the effects of different aqueous humour proteins and stressors on CTGF expression in HTM cells. HTM cells from three different donors were treated with endothelin-1, insulin-like growth factor (IGF)-1, angiotensin-II, H2 O2 and heat shock and were analysed by immunohistochemistry, real-time RT-PCR and Western blotting. Viability after H2 O2 treatment was measured in CTGF silenced HTM-N cells and their controls. Latrunculin A reduced expression of CTGF by about 50% compared to untreated HTM cells, whereas endothelin-1, IGF-1, angiotensin-II, heat shock and oxidative stress led to a significant increase. Silencing of CTGF resulted in a delayed expression of αB-crystallin and in reduced cell viability in comparison to the controls after oxidative stress. Conversely, CTGF treatment led to a higher cell viability rate after H2 O2 treatment. CTGF expression is induced by factors that have been linked to glaucoma. An increased level of CTGF appears to protect TM cells against damage induced by stress. The beneficial effect of CTGF for viability of TM cells is likely associated with the effects on increased ECM synthesis and higher contractility of the TM, thereby contributing to reduced aqueous humour outflow facility causing increased intraocular pressure.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/genética , Regulación de la Expresión Génica/genética , Malla Trabecular/metabolismo , Adulto , Anciano , Angiotensina II/farmacología , Western Blotting , Supervivencia Celular/genética , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Cristalinas/genética , Cristalinas/metabolismo , Endotelina-1/farmacología , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Calor , Humanos , Peróxido de Hidrógeno/farmacología , Factor I del Crecimiento Similar a la Insulina/farmacología , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Persona de Mediana Edad , Oxidantes/farmacología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Malla Trabecular/citología
18.
Histochem Cell Biol ; 144(5): 443-55, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26215132

RESUMEN

The stimulation of progenitor or stem cells proliferation in the retina could be a therapeutic avenue for the treatment of various ocular neurodegenerative disorders. Müller glia cells have been discussed to represent a progenitor cell population in the adult retina. In the brain, TGF-ß signaling regulates the fate of stem cells; however, its role in the vertebrate retina is unclear. We therefore investigated whether manipulation of the TGF-ß signaling pathway is sufficient to promote Müller glia cell proliferation and subsequently their trans-differentiation into retinal neurons. To this end, we used mice with heterozygous deficiency of the essential TGF-ß receptor type II or of the inhibitory protein SMAD7, in order to down- or up-regulate the activity of TGF-ß signaling, respectively. Excitotoxic damage was applied by intravitreal N-methyl-D-aspartate injection, and BrdU pulse experiments were used to label proliferative cells. Although we successfully stimulated Müller glia cell reactivity, our findings indicate that a moderate modulation of TGF-ß signaling is not sufficient to provoke Müller glia cell proliferation. Hence, TGF-ß signaling in the retina might not be the essential causative factor to maintain mammalian Müller cells in a quiescent, non-proliferative state that prevents a stem cell-like function.


Asunto(s)
N-Metilaspartato/farmacología , Neuroglía/efectos de los fármacos , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Inyecciones Intravítreas , Ratones , N-Metilaspartato/administración & dosificación , Neuroglía/metabolismo
19.
J Neurosci ; 33(35): 14246-58, 2013 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-23986258

RESUMEN

We investigated the influence of transforming growth factor-ß (TGF-ß) signaling on developmental programmed cell death in the mouse retina by direct and specific molecular targeting of TGF-ß type II receptor (TßRII) and Smad7 in retinal progenitor cells. Mice were generated carrying a conditional deletion of the TßRII in cells that originate from the inner layer of the optic cup. The animals showed a significant decrease of phosphorylated Smad3 in both the central and peripheral retina, which indicates the diminished activity of TGF-ß signaling. TßRII deficiency significantly increased the apoptotic death of retinal neurons during embryonic and postnatal development without affecting their proliferation. In contrast, treatment with TGF-ß2 inhibited cell death of retinal ganglion cells in dissociated retinal cell cultures, an effect that was blocked by inhibiting the phosphorylation of Smad3. The increase in apoptosis during development resulted in a significant reduction in the number of neurons in adult TßRII-deficient mice. The effect was most pronounced in the inner retina neurons and resulted in functional deficits as determined by electroretinography. In contrast, a conditional deletion of TGF-ß-inhibiting Smad7 in retinal neurons significantly enhanced Smad3 phosphorylation and significantly decreased apoptosis of retinal neurons in embryos and pups. Moreover, the number of retinal ganglion cells was significantly higher in Smad7-deficient mice compared with control littermates. TßRII-deficient pups showed a lower level of nerve growth factor (NGF) in its mRNA; however, higher levels were observed in Smad7-deficient pups, which strongly suggests that the protective effects of TGF-ß signaling on developmental cell death are mediated through NGF.


Asunto(s)
Apoptosis , Neuronas Retinianas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proliferación Celular , Embrión de Mamíferos , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Ratones , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Neuronas Retinianas/citología , Transducción de Señal , Proteína smad7/genética , Transcripción Genética
20.
Exp Eye Res ; 125: 41-52, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24837143

RESUMEN

Mutations in the myocilin gene (MYOC) are causative for 10% of cases with juvenile open-angle glaucoma and 3-4% of those with primary open-angle glaucoma. Myocilin is a secreted protein with relatively ill-defined matricellular properties. Despite its high expression in the eye, myocilin-deficient mice have originally been reported to have no obvious ocular phenotype. Here we revisited the ocular phenotype of myocilin-deficient mice and detected a higher number of neurons in their inner (INL) and outer (ONL) nuclear layers, as well as a higher number of retinal ganglion cells (RGC) and their axons. The increase in retinal neurons appears to be caused by a decrease in programmed developmental cell death, as apoptosis of retinal neurons between postnatal days 4 and 10 was found to be attenuated when compared to that of wildtype littermates. In contrast, when Myoc(-/-) mice were crossed with ßB1-crystallin-MYOC mice with ectopic overexpression of myocilin in the eye, no differences in developmental apoptosis, RGC number and INL thickness were observed when compared to wildtype littermates. The amounts of the anti-apoptotic Bcl-2-like protein 1 (BCL2L1, Bcl-xL) and its mRNA were increased in retinae of Myoc(-/-) mice, while lower amounts of BCL2L1 and its mRNA were detected in mixed Myoc(-/-)/ßB1-crystallin-MYOC mice. The structural differences between Myoc(-/-) mice and wildtype littermates did not result in functional differences as measured by electroretinography. Noteworthy though mixed Myoc(-/-)/ßB1-crystallin-MYOC mice with ocular overexpression of myocilin had significant cone function deficits. Myocilin appears to modulate apoptotic death of retinal neurons likely by interacting with the intrinsic apoptotic pathway.


Asunto(s)
Muerte Celular/fisiología , Proteínas del Citoesqueleto/fisiología , Proteínas del Ojo/fisiología , Glicoproteínas/fisiología , Retina/citología , Células Ganglionares de la Retina/fisiología , Animales , Apoptosis/fisiología , Proteínas del Citoesqueleto/deficiencia , Electrorretinografía , Glicoproteínas/deficiencia , Ratones Endogámicos BALB C , Ratones Transgénicos , Retina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA