Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pharm Res ; 41(1): 93-104, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37985572

RESUMEN

OBJECTIVE: To prepare polylactic acid microneedles (PLAMNs) with sustained antibacterial effect to avoid skin infection caused by traditional MNs-based biosensors. METHODS: Silver nanoparticles (AgNPs) were synthesized using an in-situ reduction process with polydopamine (PDA). PLAMNs were fabricated using the hot-melt method. A series of pressure tests and puncture experiments were conducted to confirm the physicochemical properties of PLAMNs. Then AgNPs were modified on the surface of PLAMNs through in-situ reduction of PDA, resulting in the formation of PLAMNs@PDA-AgNPs. The in vitro antibacterial efficacy of PLAMNs@PDA-AgNPs was evaluated using agar diffusion assays and bacterial liquid co-culture approach. Wound healing and simulated long-term application were performed to assess the in vivo antibacterial effectiveness of PLAMNs@PDA-AgNPs. RESULTS: The MNs array comprised 169 tiny needle tips in pyramidal rows. Strength and puncture tests confirmed a 100% puncture success rate for PLAMNs on isolated rat skin and tin foil. SEM analysis revealed the integrity of PLAMNs@PDA-AgNPs with the formation of new surface substances. EDS analysis indicated the presence of silver elements on the surface of PLAMNs@PDA-AgNPs, with a content of 14.44%. Transepidermal water loss (TEWL) testing demonstrated the rapid healing of micro-pores created by PLAMNs@PDA-AgNPs, indicating their safety. Both in vitro and in vivo tests confirmed antibacterial efficacy of PLAMNs@PDA-AgNPs. CONCLUSIONS: In conclusion, the sustained antibacterial activity exhibited by PLAMNs@PDA-AgNPs offers a promising solution for addressing skin infections associated with MN applications, especially when compared to traditional MN-based biosensors. This advancement offers significant potential for the field of MN technology.


Asunto(s)
Nanopartículas del Metal , Poliésteres , Plata , Ratas , Animales , Plata/farmacología , Plata/química , Nanopartículas del Metal/química , Staphylococcus aureus , Antibacterianos/farmacología , Antibacterianos/química , Pruebas de Sensibilidad Microbiana
2.
Graefes Arch Clin Exp Ophthalmol ; 256(11): 2113-2125, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30215097

RESUMEN

PURPOSE: To create new immunodeficient Royal College of Surgeons (RCS) rats by introducing the defective MerTK gene into athymic nude rats. METHODS: Female homozygous RCS (RCS-p+/RCS-p+) and male nude rats (Hsd:RH-Foxn1mu, mutation in the foxn1 gene; no T cells) were crossed to produce heterozygous F1 progeny. Double homozygous F2 progeny obtained by crossing the F1 heterozygotes was identified phenotypically (hair loss) and genotypically (RCS-p+ gene determined by PCR). Retinal degenerative status was confirmed by optical coherence tomography (OCT) imaging, electroretinography (ERG), optokinetic (OKN) testing, superior colliculus (SC) electrophysiology, and by histology. The effect of xenografts was assessed by transplantation of human embryonic stem cell-derived retinal pigment epithelium (hESC-RPE) and human-induced pluripotent stem cell-derived RPE (iPS-RPE) into the eye. Morphological analysis was conducted based on hematoxylin and eosin (H&E) and immunostaining. Age-matched pigmented athymic nude rats were used as control. RESULTS: Approximately 6% of the F2 pups (11/172) were homozygous for RCS-p+ gene and Foxn1mu gene. Homozygous males crossed with heterozygous females resulted in 50% homozygous progeny for experimentation. OCT imaging demonstrated significant loss of retinal thickness in homozygous rats. H&E staining showed photoreceptor thickness reduced to 1-3 layers at 12 weeks of age. Progressive loss of visual function was evidenced by OKN testing, ERG, and SC electrophysiology. Transplantation experiments demonstrated survival of human-derived cells and absence of apparent immune rejection. CONCLUSIONS: This new rat animal model developed by crossing RCS rats and athymic nude rats is suitable for conducting retinal transplantation experiments involving xenografts.


Asunto(s)
Modelos Animales de Enfermedad , Células Madre Embrionarias Humanas/trasplante , Síndromes de Inmunodeficiencia/terapia , Células Madre Pluripotentes Inducidas/trasplante , Distrofias Retinianas/terapia , Epitelio Pigmentado de la Retina/trasplante , Animales , Supervivencia Celular , Electrorretinografía , Femenino , Técnicas de Genotipaje , Supervivencia de Injerto/fisiología , Células Madre Embrionarias Humanas/fisiología , Humanos , Síndromes de Inmunodeficiencia/diagnóstico , Síndromes de Inmunodeficiencia/fisiopatología , Células Madre Pluripotentes Inducidas/fisiología , Masculino , Fenotipo , Ratas , Ratas Desnudas , Retina/fisiopatología , Distrofias Retinianas/diagnóstico , Distrofias Retinianas/fisiopatología , Epitelio Pigmentado de la Retina/fisiología , Tomografía de Coherencia Óptica , Tirosina Quinasa c-Mer/genética
3.
Am J Pathol ; 186(4): 859-73, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26878210

RESUMEN

Subretinal fibrosis is an end stage of neovascular age-related macular degeneration, characterized by fibrous membrane formation after choroidal neovascularization. An initial step of the pathogenesis is an epithelial-mesenchymal transition (EMT) of retinal pigment epithelium cells. αB-crystallin plays multiple roles in age-related macular degeneration, including cytoprotection and angiogenesis. However, the role of αB-crystallin in subretinal EMT and fibrosis is unknown. Herein, we showed attenuation of subretinal fibrosis after regression of laser-induced choroidal neovascularization and a decrease in mesenchymal retinal pigment epithelium cells in αB-crystallin knockout mice compared with wild-type mice. αB-crystallin was prominently expressed in subretinal fibrotic lesions in mice. In vitro, overexpression of αB-crystallin induced EMT, whereas suppression of αB-crystallin induced a mesenchymal-epithelial transition. Transforming growth factor-ß2-induced EMT was further enhanced by overexpression of αB-crystallin but was inhibited by suppression of αB-crystallin. Silencing of αB-crystallin inhibited multiple fibrotic processes, including cell proliferation, migration, and fibronectin production. Bone morphogenetic protein 4 up-regulated αB-crystallin, and its EMT induction was inhibited by knockdown of αB-crystallin. Furthermore, inhibition of αB-crystallin enhanced monotetraubiquitination of SMAD4, which can impair its nuclear localization. Overexpression of αB-crystallin enhanced nuclear translocation and accumulation of SMAD4 and SMAD5. Thus, αB-crystallin is an important regulator of EMT, acting as a molecular chaperone for SMAD4 and as its potential therapeutic target for preventing subretinal fibrosis development in neovascular age-related macular degeneration.


Asunto(s)
Neovascularización Coroidal/metabolismo , Transición Epitelial-Mesenquimal/genética , Fibrosis/metabolismo , Degeneración Macular/patología , Epitelio Pigmentado de la Retina/metabolismo , Cadena B de alfa-Cristalina/metabolismo , Animales , Neovascularización Coroidal/genética , Fibronectinas/metabolismo , Humanos , Degeneración Macular/genética , Masculino , Ratones Noqueados , Epitelio Pigmentado de la Retina/patología , Cadena B de alfa-Cristalina/genética
4.
Proc Natl Acad Sci U S A ; 111(47): 16754-9, 2014 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-25385631

RESUMEN

The retinal pigmented epithelial (RPE) layer is one of the major ocular tissues affected by oxidative stress and is known to play an important role in the etiology of age-related macular degeneration (AMD), the major cause of blinding in the elderly. In the present study, sulindac, a nonsteroidal antiinflammatory drug (NSAID), was tested for protection against oxidative stress-induced damage in an established RPE cell line (ARPE-19). Besides its established antiinflammatory activity, sulindac has previously been shown to protect cardiac tissue against ischemia/reperfusion damage, although the exact mechanism was not elucidated. As shown here, sulindac can also protect RPE cells from chemical oxidative damage or UV light by initiating a protective mechanism similar to what is observed in ischemic preconditioning (IPC) response. The mechanism of protection appears to be triggered by reactive oxygen species (ROS) and involves known IPC signaling components such as PKG and PKC epsilon in addition to the mitochondrial ATP-sensitive K(+) channel. Sulindac induced iNOS and Hsp70, late-phase IPC markers in the RPE cells. A unique feature of the sulindac protective response is that it involves activation of the peroxisome proliferator-activated receptor alpha (PPAR-α). We have also used low-passage human fetal RPE and polarized primary fetal RPE cells to validate the basic observation that sulindac can protect retinal cells against oxidative stress. These findings indicate a mechanism for preventing oxidative stress in RPE cells and suggest that sulindac could be used therapeutically for slowing the progression of AMD.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , PPAR alfa/fisiología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Sulindac/farmacología , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Oxidativo
5.
Graefes Arch Clin Exp Ophthalmol ; 254(8): 1553-1565, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27335025

RESUMEN

PURPOSE: A subretinal implant termed CPCB-RPE1 is currently being developed to surgically replace dystrophic RPE in patients with dry age-related macular degeneration (AMD) and severe vision loss. CPCB-RPE1 is composed of a terminally differentiated, polarized human embryonic stem cell-derived RPE (hESC-RPE) monolayer pre-grown on a biocompatible, mesh-supported submicron parylene C membrane. The objective of the present delivery study was to assess the feasibility and 1-month safety of CPCB-RPE1 implantation in Yucatán minipigs, whose eyes are similar to human eyes in size and gross retinal anatomy. METHODS: This was a prospective, partially blinded, randomized study in 14 normal-sighted female Yucatán minipigs (aged 2 months, weighing 24-35 kg). Surgeons were blinded to the randomization codes and postoperative and post-mortem assessments were performed in a blinded manner. Eleven minipigs received CPCB-RPE1 while three control minipigs underwent sham surgery that generated subretinal blebs. All animals except two sham controls received combined local (Ozurdex™ dexamethasone intravitreal implant) and systemic (tacrolimus) immunosuppression or local immunosuppression alone. Correct placement of the CPCB-RPE1 implant was assessed by in vivo optical coherence tomography and post-mortem histology. hESC-RPE cells were identified using immunohistochemistry staining for TRA-1-85 (a human marker) and RPE65 (an RPE marker). As the study results of primary interest were nonnumerical no statistical analysis or tests were conducted. RESULTS: CPCB-RPE1 implants were reliably placed, without implant breakage, in the subretinal space of the minipig eye using surgical techniques similar to those that would be used in humans. Histologically, hESC-RPE cells were found to survive as an intact monolayer for 1 month based on immunohistochemistry staining for TRA-1-85 and RPE65. CONCLUSIONS: Although inconclusive regarding the necessity or benefit of systemic or local immunosuppression, our study demonstrates the feasibility and safety of CPCB-RPE1 subretinal implantation in a comparable animal model and provides an encouraging starting point for human studies.


Asunto(s)
Células Madre Embrionarias Humanas/trasplante , Degeneración Macular/cirugía , Epitelio Pigmentado de la Retina/trasplante , Trasplante de Células Madre/métodos , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Estudios de Factibilidad , Femenino , Humanos , Degeneración Macular/diagnóstico , Estudios Prospectivos , Epitelio Pigmentado de la Retina/citología , Porcinos , Porcinos Enanos , Tomografía de Coherencia Óptica , Resultado del Tratamiento
6.
Cytokine ; 71(2): 394-6, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25496702

RESUMEN

Retinal pigmented epithelium (RPE) secretes transforming growth factor beta 1 and 2 (TGF-ß1 and -ß2) cytokines involved in fibrosis, immune privilege, and proliferative vitreoretinopathy (PVR). Since RPE cell polarity may be altered in various disease conditions including PVR and age-related macular degeneration, we determined levels of TGF-ß from polarized human RPE (hRPE) and human stem cell derived RPE (hESC-RPE) as compared to nonpolarized cells. TGF-ß2 was the predominant isoform in all cell culture conditions. Nonpolarized cells secreted significantly more TGF-ß2 supporting the contention that loss of polarity of RPE in PVR leads to rise of intravitreal TGF-ß2. Active TGF-ß2, secreted mainly from apical side of polarized RPE, represented 6-10% of total TGF-ß2. In conclusion, polarity is an important determinant of TGF-ß2 secretion in RPE. Low levels of apically secreted active TGF-ß2 may play a role in the normal physiology of the subretinal space. Comparable secretion of TGF-ß from polarized hESC-RPE and hRPE supports the potential for hESC-RPE in RPE replacement therapies.


Asunto(s)
Retina/citología , Epitelio Pigmentado de la Retina/citología , Células Madre/citología , Factor de Crecimiento Transformador beta2/metabolismo , Trasplante de Células , Células Cultivadas , Células Madre Embrionarias/citología , Humanos , Isoformas de Proteínas/metabolismo , Retina/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Vitreorretinopatía Proliferativa/patología
7.
Curr Pharm Biotechnol ; 25(4): 477-487, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37345239

RESUMEN

Background: Cinnamic acid and its derivatives have gained significant attention in recent medicinal research due to their broad spectrum of pharmacological properties. However, the effects of these compounds on xanthine oxidase (XO) have not been systematically investigated, and the inhibitory mechanism remains unclear.

Objectives: The objective of this study was to screen 18 compounds and identify the XO inhibitor with the strongest inhibitory effect. Furthermore, we aimed to study the inhibitory mechanism of the identified compound.

Methods: The effects of the inhibitors on XO were evaluated using kinetic analysis, docking simulations, and in vivo study. Among the compounds tested, 4-NA was discovered as the first XO inhibitor and exhibited the most potent inhibitory effects, with an IC50 value of 23.02 ± 0.12 µmol/L. The presence of the nitro group in 4-NA was found to be essential for enhancing XO inhibition. The kinetic study revealed that 4-NA inhibited XO in a reversible and noncompetitive manner. Moreover, fluorescence spectra analysis demonstrated that 4-NA could spontaneously form complexes with XO, referred to as 4-NA-XO complexes, with the negative values of △H and ΔS.

Results: This suggests that hydrogen bonds and van der Waals forces play crucial roles in the binding process. Molecular docking studies further supported the kinetic analysis and provided insight into the optimal binding conformation, indicating that 4-NA is located at the bottom outside the catalytic center through the formation of three hydrogen bonds. Furthermore, animal studies confirmed that the inhibitory effects of 4-NA on XO resulted in a significant reduction of serum uric acid level in hyperuricemia mice.

Conclusion: This work elucidates the mechanism of 4-NA inhibiting XO, paving the way for the development of new XO inhibitors.

.


Asunto(s)
Hiperuricemia , Xantina Oxidasa , Ratones , Animales , Simulación del Acoplamiento Molecular , Xantina Oxidasa/química , Xantina Oxidasa/metabolismo , Ácido Úrico , Cinética , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química
8.
Pharmaceutics ; 15(6)2023 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-37376179

RESUMEN

Microneedles (MNs) have recently garnered extensive interest concerning direct interstitial fluid (ISF) extraction or their integration into medical devices for continuous biomarker monitoring, owing to their advantages of painlessness, minimal invasiveness, and ease of use. However, micropores created by MN insertion may provide pathways for bacterial infiltration into the skin, causing local or systemic infection, especially with long-term in situ monitoring. To address this, we developed a novel antibacterial sponge MNs (SMNs@PDA-AgNPs) by depositing silver nanoparticles (AgNPs) on polydopamine (PDA)-coated SMNs. The physicochemical properties of SMNs@PDA-AgNPs were characterized regarding morphology, composition, mechanical strength, and liquid absorption capacity. The antibacterial effects were evaluated and optimized through agar diffusion assays in vitro. Wound healing and bacterial inhibition were further examined in vivo during MN application. Finally, the ISF sampling ability and biosafety of SMNs@PDA-AgNPs were assessed in vivo. The results demonstrate that antibacterial SMNs enable direct ISF extraction while preventing infection risks. SMNs@PDA-AgNPs could potentially be used for direct sampling or combined with medical devices for real-time diagnosis and management of chronic diseases.

9.
IET Nanobiotechnol ; 17(4): 368-375, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37153957

RESUMEN

Ellagic acid (EA), which is widely distributed in many foods, has been found to possess inhibitory activity against xanthine oxidase (XO). However, there is ongoing debate about the difference in XO inhibitory activity between EA and allopurinol. Additionally, the inhibitory kinetics and mechanism of EA on XO are still unclear. Herein, the authors systematically studied the inhibitory effects of EA on XO. The authors' findings showed that EA is a reversible inhibitor with mixed-type inhibition, and its inhibitory activity is weaker than allopurinol. Fluorescence quenching experiments suggested that the generation of EA-XO complex was exothermic and spontaneous. In silico analysis further confirmed that EA entered the XO catalytic centre. Furthermore, the authors verified the anti-hyperuricemia effect of EA in vivo. This study elucidates the inhibition kinetics and mechanism of EA on XO, and lays a theoretical foundation for the further development of drugs and functional foods containing EA for the treatment of hyperuricemia.


Asunto(s)
Alopurinol , Hiperuricemia , Humanos , Alopurinol/farmacología , Alopurinol/uso terapéutico , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Xantina Oxidasa/metabolismo , Xantina Oxidasa/uso terapéutico , Ácido Elágico/farmacología , Ácido Elágico/uso terapéutico , Cinética , Hiperuricemia/tratamiento farmacológico
10.
Angiogenesis ; 15(2): 213-27, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22392094

RESUMEN

Bone morphorgenetic protein (BMP)-4 has been shown to play a pivotal role in eye development; however, its role in mature retina or ocular angiogenic diseases is unclear. Activating downstream Smad signaling, BMP4 can be either pro-angiogenic or anti-angiogenic, depending on the context of cell types and associated microenvironment. In this study, we generated transgenic mice over-expressing BMP4 in retinal pigment epithelial (RPE) cells (Vmd2-Bmp4 Tg mice), and used the laser-induced choroidal neovascularization (CNV) model to study the angiogenic properties of BMP4 in adult eyes. Vmd2-Bmp4 Tg mice displayed normal retinal histology at 10 weeks of age when compared with age-matched wildtype mice. Over-expression of BMP4 in RPE in the transgenic mice was confirmed by real-time PCR and immunostaining. Elevated levels of Smad1,5 phosphorylation were found in BMP4 transgenic mice compared to wildype mice. Over-expression of BMP4 was associated with less severe CNV as characterized by fluorescein angiography, CNV volume measurement and histology. While control mice showed increased levels of vascular endothelial growth factor (VEGF) and matrix metalloproteinase (MMP)-9 after laser injury, Vmd2-Bmp4 Tg showed no increase in either VEGF or MMP-9. Further, we found that TNF-induced MMP-9 secretion in vitro was reduced by pretreatment of RPE cells with BMP4. The inhibition of MMP-9 was Smad-dependent because BMP4 failed to repress TNF-induced MMP-9 expression when Smad1,5 was silenced by siRNA. In summary, our studies identified an anti-angiogenic role for BMP4 in laser-induced CNV, mediated by direct inhibition of MMP-9 and indirect inhibition of VEGF.


Asunto(s)
Proteína Morfogenética Ósea 4/biosíntesis , Neovascularización Coroidal/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Bestrofinas , Proteína Morfogenética Ósea 4/genética , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Neovascularización Coroidal/etiología , Neovascularización Coroidal/genética , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Regulación Enzimológica de la Expresión Génica/genética , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Rayos Láser/efectos adversos , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Transgénicos , Epitelio Pigmentado de la Retina/patología , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
11.
FASEB J ; 25(7): 2221-33, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21411747

RESUMEN

Bone morphogenetic protein-4 (BMP4) may be involved in the molecular switch that determines which late form of age-related macular degeneration (AMD) an individual develops. BMP4 expression is high in retinal pigment epithelium (RPE) cells in late, dry AMD patients, while BMP4 expression is low in the wet form of the disease, characterized by choroidal neovascularization (CNV). Here, we sought to determine the mechanism by which BMP4 is down-regulated in CNV. BMP4 expression was decreased within laser-induced CNV lesions in mice at a time when tumor necrosis factor (TNF) expression was high (7 d postlaser) and was reexpressed in RPE when TNF levels declined (14 d postlaser). We found that TNF, an important angiogenic stimulus, significantly down-regulates BMP4 expression in cultured human fetal RPE cells, ARPE-19 cells, and RPE cells in murine posterior eye cup explants. We identified two specificity protein 1 (Sp1) binding sites in the BMP4 promoter that are required for basal expression of BMP4 and its down-regulation by TNF. Through c-Jun NH(2)-terminal kinase (JNK) activation, TNF modulates Sp1 phosphorylation, thus decreasing its affinity to the BMP4 promoter. The down-regulation of BMP4 expression by TNF in CNV and mechanisms established might be useful for defining novel targets for AMD therapy.


Asunto(s)
Proteína Morfogenética Ósea 4/genética , Regulación de la Expresión Génica , Degeneración Macular/genética , Factor de Necrosis Tumoral alfa/genética , Animales , Sitios de Unión/genética , Western Blotting , Proteína Morfogenética Ósea 4/metabolismo , Línea Celular , Células Cultivadas , Neovascularización Coroidal/etiología , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Rayos Láser/efectos adversos , Degeneración Macular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción Sp1/metabolismo , Transcripción Genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
12.
Biomed Microdevices ; 14(4): 659-67, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22391881

RESUMEN

In this work, a mesh-supported submicron parylene-C membrane (MSPM) is proposed as an artificial Bruch's membrane for the therapy of age-related macular degeneration (AMD). Any artificial Bruch's membrane must first satisfy two important requirements. First, it should be as permeable as healthy human Bruch's membrane to support nutrients transportation. Secondly, it should be able to support the adherence and proliferation of retinal pigment epithelial (RPE) cells with in vivo-like morphologies and functions. Although parylene-C is widely used as a barrier layer in many biomedical applications, it is found that parylene-C membranes with submicron thickness are semipermeable to macromolecules. We first measure the permeability of submicron parylene-C and find that 0.15-0.30 µm parylene-C has similar permeability to healthy human Bruch's membranes. Blind-well perfusion cell viability experiments further demonstrate that nutrients and macromolecules can diffuse across 0.30 µm parylene-C to nourish the cells. A mesh-supported submicron parylene-C membrane (MSPM) structure is design to enhance the mechanical strength of the substrate. In vitro cells culture on the MSPM (with 0.30 µm ultrathin parylene-C) shows that H9-RPE cells are able to adhere, proliferate, form epithelial monolayer with tight intracellular junctions, and become well-polarized with microvilli, which exhibit similar characteristics to RPE cells in vivo. These studies have demonstrated the potential of the MSPM as an artificial Bruch's membrane for RPE cell transplantation.


Asunto(s)
Técnicas de Cultivo de Célula/instrumentación , Membranas Artificiales , Polímeros/química , Epitelio Pigmentado de la Retina/citología , Xilenos/química , Línea Celular , Proliferación Celular , Supervivencia Celular , Difusión , Diseño de Equipo , Humanos , Fenómenos Mecánicos , Peso Molecular , Permeabilidad
13.
Ophthalmic Res ; 48(4): 186-91, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22868580

RESUMEN

OBJECTIVE: To evaluate the feasibility of a new technique for the implantation of ultrathin substrates containing stem cell-derived retinal pigment epithelium (RPE) cells into the subretinal space of retina-degenerate Royal College of Surgeon (RCS) rats. METHODS: A platform device was used for the implantation of 4-µm-thick parylene substrates containing a monolayer of human embryonic stem cell-derived RPE (hESC-RPE). Normal Copenhagen rats (n = 6) and RCS rats (n = 5) were used for the study. Spectral-domain optical coherence tomography (SD-OCT) scanning and histological examinations were performed to confirm placement location of the implant. hESC-RPE cells attached to the substrate before and after implantation were evaluated using standard cell counting techniques. RESULTS: SD-OCT scanning and histological examination revealed that the substrates were precisely placed in the rat's subretinal space. The hESC-RPE cell monolayer that covered the surface of the substrate was found to be intact after implantation. Cell counting data showed that less than 2% of cells were lost from the substrate due to the implantation procedure (preimplantation count 2,792 ± 74.09 cells versus postimplantation count 2,741 ± 62.08 cells). Detailed microscopic examination suggested that the cell loss occurred mostly along the edges of the implant. CONCLUSION: With the help of this platform device, it is possible to implant ultrathin substrates containing an RPE monolayer into the rat's subretinal space. This technique can be a useful approach for stem cell-based tissue bioengineering techniques in retinal transplantation research.


Asunto(s)
Células Madre Embrionarias/citología , Polímeros , Distrofias Retinianas/terapia , Epitelio Pigmentado de la Retina/trasplante , Trasplante de Células Madre , Ingeniería de Tejidos , Andamios del Tejido , Xilenos , Animales , Recuento de Células , Estudios de Factibilidad , Humanos , Ratas , Ratas Mutantes , Retina/patología , Distrofias Retinianas/diagnóstico , Tomografía de Coherencia Óptica
14.
J Vis Exp ; (190)2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36533846

RESUMEN

Retinal degeneration, such as age-related macular degeneration (AMD), is a leading cause of blindness worldwide. A myriad of approaches have been undertaken to develop regenerative medicine-based therapies for AMD, including stem cell-based therapies. Rodents as animal models for retinal degeneration are a foundation for translational research, due to the broad spectrum of strains that develop retinal degeneration diseases at different stages. However, mimicking human therapeutic delivery of subretinal implants in rodents is challenging, due to anatomical differences such as lens size and vitreous volume. This surgical protocol aims to provide a guided method for transplanting implants into the subretinal space in rats. A user-friendly comprehensive description of the critical steps has been included. This protocol has been developed as a cost-efficient surgical procedure for reproducibility across different preclinical studies in rats. Proper miniaturization of a human-sized implant is required prior to conducting the surgical experiment, which includes adjustments to the dimensions of the implant. An external approach is used instead of an intravitreal procedure to deliver the implant to the subretinal space. Using a small sharp needle, a scleral incision is performed in the temporal superior quadrant, followed by paracentesis to reduce intraocular pressure, thereby minimizing resistance during the surgical implantation. Next, a balanced salt solution (BSS) injection through the incision is carried out to achieve focal retinal detachment (RD). Lastly, insertion and visualization of the implant into the subretinal space are conducted. Post-operative assessment of the subretinal placement of the implant includes imaging by spectral domain optical coherence tomography (SD-OCT). Imaging follow-ups ascertain the subretinal stability of the implant, before the eyes are harvested and fixated for histological analysis.


Asunto(s)
Degeneración Macular , Degeneración Retiniana , Humanos , Ratas , Animales , Degeneración Retiniana/cirugía , Degeneración Retiniana/patología , Reproducibilidad de los Resultados , Modelos Animales de Enfermedad , Degeneración Macular/terapia , Tomografía de Coherencia Óptica/métodos
15.
Stem Cell Reports ; 17(3): 448-458, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35120620

RESUMEN

Cell-based therapies face challenges, including poor cell survival, immune rejection, and integration into pathologic tissue. We conducted an open-label phase 1/2a clinical trial to assess the safety and preliminary efficacy of a subretinal implant consisting of a polarized monolayer of allogeneic human embryonic stem cell-derived retinal pigmented epithelium (RPE) cells in subjects with geographic atrophy (GA) secondary to dry age-related macular degeneration. Postmortem histology from one subject with very advanced disease shows the presence of donor RPE cells 2 years after implantation by immunoreactivity for RPE65 and donor-specific human leukocyte antigen (HLA) class I molecules. Markers of RPE cell polarity and phagocytosis suggest donor RPE function. Further histologic examination demonstrated CD34+ structures beneath the implant and CD4+, CD68+, and FoxP3+ cells in the tissue. Despite significant donor-host HLA mismatch, no clinical signs of retinitis, vitreitis, vasculitis, choroiditis, or serologic immune response were detected in the deceased subject or any other subject in the study. Subretinally implanted, HLA-mismatched donor RPE cells survive, express functional markers, and do not elicit clinically detectable intraocular inflammation or serologic immune responses even without long-term immunosuppression.


Asunto(s)
Atrofia Geográfica , Degeneración Macular , Prótesis e Implantes , Atrofia Geográfica/terapia , Células Madre Embrionarias Humanas/patología , Humanos , Degeneración Macular/patología , Degeneración Macular/terapia , Prótesis e Implantes/efectos adversos , Epitelio Pigmentado de la Retina/patología
16.
Cells ; 10(11)2021 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-34831174

RESUMEN

Retinal pigment epithelium (RPE) replacement therapy is evolving as a feasible approach to treat age-related macular degeneration (AMD). In many preclinical studies, RPE cells are transplanted as a cell suspension into immunosuppressed animal eyes and transplant effects have been monitored only short-term. We investigated the long-term effects of human Induced pluripotent stem-cell-derived RPE (iPSC-RPE) transplants in an immunodeficient Royal College of Surgeons (RCS) rat model, in which RPE dysfunction led to photoreceptor degeneration. iPSC-RPE cultured as a polarized monolayer on a nanoengineered ultrathin parylene C scaffold was transplanted into the subretinal space of 28-day-old immunodeficient RCS rat pups and evaluated after 1, 4, and 11 months. Assessment at early time points showed good iPSC-RPE survival. The transplants remained as a monolayer, expressed RPE-specific markers, performed phagocytic function, and contributed to vision preservation. At 11-months post-implantation, RPE survival was observed in only 50% of the eyes that were concomitant with vision preservation. Loss of RPE monolayer characteristics at the 11-month time point was associated with peri-membrane fibrosis, immune reaction through the activation of macrophages (CD 68 expression), and the transition of cell fate (expression of mesenchymal markers). The overall study outcome supports the therapeutic potential of RPE grafts despite the loss of some transplant benefits during long-term observations.


Asunto(s)
Células Madre Pluripotentes Inducidas/trasplante , Epitelio Pigmentado de la Retina/trasplante , Animales , Biomarcadores/metabolismo , Humanos , Implantes Experimentales , Luz , Polímeros , Ratas , Colículos Superiores/efectos de la radiación , Análisis de Supervivencia , Visión Ocular/efectos de la radiación , Xilenos
17.
Front Neurosci ; 15: 752958, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34764853

RESUMEN

End-stage age-related macular degeneration (AMD) and retinitis pigmentosa (RP) are two major retinal degenerative (RD) conditions that result in irreversible vision loss. Permanent eye damage can also occur in battlefields or due to accidents. This suggests there is an unmet need for developing effective strategies for treating permanent retinal damages. In previous studies, co-grafted sheets of fetal retina with its retinal pigment epithelium (RPE) have demonstrated vision improvement in rat retinal disease models and in patients, but this has not yet been attempted with stem-cell derived tissue. Here we demonstrate a cellular therapy for irreversible retinal eye injuries using a "total retina patch" consisting of retinal photoreceptor progenitor sheets and healthy RPE cells on an artificial Bruch's membrane (BM). For this, retina organoids (ROs) (cultured in suspension) and polarized RPE sheets (cultured on an ultrathin parylene substrate) were made into a co-graft using bio-adhesives [gelatin, growth factor-reduced matrigel, and medium viscosity (MVG) alginate]. In vivo transplantation experiments were conducted in immunodeficient Royal College of Surgeons (RCS) rats at advanced stages of retinal degeneration. Structural reconstruction of the severely damaged retina was observed based on histological assessments and optical coherence tomography (OCT) imaging. Visual functional assessments were conducted by optokinetic behavioral testing and superior colliculus electrophysiology. Long-term survival of the co-graft in the rat subretinal space and improvement in visual function were observed. Immunohistochemistry showed that co-grafts grew, generated new photoreceptors and developed neuronal processes that were integrated into the host retina. This novel approach can be considered as a new therapy for complete replacement of a degenerated retina.

18.
Sci Rep ; 11(1): 6286, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33737600

RESUMEN

Age-related macular degeneration (AMD) is the primary cause of blindness in adults over 60 years of age, and clinical trials are currently assessing the therapeutic potential of retinal pigmented epithelial (RPE) cell monolayers on implantable scaffolds to treat this disease. However, challenges related to the culture, long-term storage, and long-distance transport of such implants currently limit the widespread use of adherent RPE cells as therapeutics. Here we report a xeno-free protocol to cryopreserve a confluent monolayer of clinical-grade, human embryonic stem cell-derived RPE cells on a parylene scaffold (REPS) that yields viable, polarized, and functional RPE cells post-thaw. Thawed cells exhibit ≥ 95% viability, have morphology, pigmentation, and gene expression characteristic of mature RPE cells, and secrete the neuroprotective protein, pigment epithelium-derived factor (PEDF). Stability under liquid nitrogen (LN2) storage has been confirmed through one year. REPS were administered immediately post-thaw into the subretinal space of a mammalian model, the Royal College of Surgeons (RCS)/nude rat. Implanted REPS were assessed at 30, 60, and 90 days post-implantation, and thawed cells demonstrate survival as an intact monolayer on the parylene scaffold. Furthermore, immunoreactivity for the maturation marker, RPE65, significantly increased over the post-implantation period in vivo, and cells demonstrated functional attributes similar to non-cryopreserved controls. The capacity to cryopreserve adherent cellular therapeutics permits extended storage and stable transport to surgical sites, enabling broad distribution for the treatment of prevalent diseases such as AMD.


Asunto(s)
Criopreservación/métodos , Células Epiteliales/trasplante , Degeneración Macular/terapia , Epitelio Pigmentado de la Retina/trasplante , Manejo de Especímenes/métodos , Trasplante de Células Madre/métodos , Animales , Diferenciación Celular , Línea Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas del Ojo/metabolismo , Células Madre Embrionarias Humanas/citología , Humanos , Factores de Crecimiento Nervioso/metabolismo , Polímeros , Ratas , Ratas Desnudas , Medicina Regenerativa/métodos , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/metabolismo , Serpinas/metabolismo , Andamios del Tejido , Resultado del Tratamiento , Xilenos
19.
Transl Vis Sci Technol ; 8(3): 31, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31171998

RESUMEN

PURPOSE: To characterize histologic changes in the optic nerve and the retina of an end-stage retinitis pigmentosa (RP) patient after long-term implantation with the Argus II retinal prosthesis system. METHODS: Serial cross sections from the patient's both eyes were collected postmortem 6 years after implantation. Optic nerve from both eyes were morphometrically analyzed and compared. Retina underneath and outside the array was analyzed and compared with corresponding regions in the fellow eye. RESULTS: Although the optic nerve of the implant eye demonstrated significantly more overall atrophy than the fellow eye (P < 0.01), the temporal quadrant that retinotopically corresponded to the location of the array did not show additional damage. The total neuron count of the macular area was not significantly different between the two eyes, but the tack locations and their adjacent areas showed significantly fewer neurons than other perimacular areas. There was an increased expression of glial fibrillary acidic protein (GFAP) throughout the retina in the implant eye versus the fellow eye, but there was no significant difference in the cellular retinaldehyde-binding protein (CRALBP) expression. Except for the revision tack site, no significant increase of inflammatory reaction was detected in the implant eye. CONCLUSION: Long-term implantation and electrical stimulation with an Argus II retinal prosthesis system did not result in significant tissue damage that could be detected by a morphometric analysis. TRANSLATIONAL RELEVANCE: This study supports the long-term safety of the Argus II device and encourages further development of bioelectronics devices at the retina-machine interface.

20.
Biochem Biophys Res Commun ; 367(1): 1-6, 2008 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-18086559

RESUMEN

Leucine-rich amelogenin peptide (LRAP), an alternatively spliced amelogenin protein, possesses a signaling property shown to induce osteogenic differentiation. In the current study, we detected LRAP expression during osteogenesis of wild-type (WT) embryonic stem (ES) cells and observed the absence of LRAP expression in amelogenin-null (KO) ES cells. We explored the signaling effect of LRAP on wild-type ES cells, and the ability of LRAP to rescue the impaired osteogenesis phenotype observed in KO ES cells. Our data indicate that LRAP treatment of WT and KO ES cells induces a significant increase in mineral matrix formation, and significant increases in bone sialoprotein and osterix gene expression. In addition, the amelogenin KO phenotype is partially rescued by the addition of exogenous LRAP. These data suggest a unique function of LRAP during ES cell differentiation along osteogenic lineage.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas del Esmalte Dental/farmacología , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Animales , Secuencia de Bases , Regeneración Ósea/fisiología , Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocalcina/metabolismo , Osteogénesis/fisiología , Fenotipo , Sialoglicoproteínas/biosíntesis , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA