Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.095
Filtrar
1.
Biomaterials ; 313: 122770, 2025 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-39226653

RESUMEN

Major advances have been made in utilizing human-induced pluripotent stem cells (hiPSCs) for regenerative medicine. Nevertheless, the delivery and integration of hiPSCs into target tissues remain significant challenges, particularly in the context of retinal ganglion cell (RGC) restoration. In this study, we introduce a promising avenue for providing directional guidance to regenerated cells in the retina. First, we developed a technique for construction of gradient interfaces based on functionalized conductive polymers, which could be applied with various functionalized ehthylenedioxythiophene (EDOT) monomers. Using a tree-shaped channel encapsulated with a thin PDMS and a specially designed electrochemical chamber, gradient flow generation could be converted into a functionalized-PEDOT gradient film by cyclic voltammetry. The characteristics of the successfully fabricated gradient flow and surface were analyzed using fluorescent labels, time of flight secondary ion mass spectrometry (TOF-SIMS), and X-ray photoelectron spectroscopy (XPS). Remarkably, hiPSC-RGCs seeded on PEDOT exhibited improvements in neurite outgrowth, axon guidance and neuronal electrophysiology measurements. These results suggest that our novel gradient PEDOT may be used with hiPSC-based technologies as a potential biomedical engineering scaffold for functional restoration of RGCs in retinal degenerative diseases and optic neuropathies.


Asunto(s)
Células Madre Pluripotentes Inducidas , Polímeros , Células Ganglionares de la Retina , Humanos , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/citología , Células Madre Pluripotentes Inducidas/citología , Polímeros/química , Orientación del Axón , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Propiedades de Superficie , Conductividad Eléctrica , Factores de Crecimiento Nervioso/metabolismo , Axones/metabolismo , Axones/fisiología
2.
J Cell Biol ; 223(12)2024 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-39352499

RESUMEN

Successful axonal regeneration following injury requires the effective allocation of energy. How axons withstand the initial disruption in mitochondrial energy production caused by the injury and subsequently initiate regrowth is poorly understood. Transcriptomic data showed increased expression of glycolytic genes after optic nerve crush in retinal ganglion cells with the co-deletion of Pten and Socs3. Using retinal cultures in a multicompartment microfluidic device, we observed increased regrowth and enhanced mitochondrial trafficking in the axons of Pten and Socs3 co-deleted neurons. While wild-type axons relied on mitochondrial metabolism, after injury, in the absence of Pten and Socs3, energy production was supported by local glycolysis. Specific inhibition of lactate production hindered injury survival and the initiation of regrowth while slowing down glycolysis upstream impaired regrowth initiation, axonal elongation, and energy production. Together, these observations reveal that glycolytic ATP, combined with sustained mitochondrial transport, is essential for injury-induced axonal regrowth, providing new insights into the metabolic underpinnings of axonal regeneration.


Asunto(s)
Axones , Glucólisis , Mitocondrias , Regeneración Nerviosa , Células Ganglionares de la Retina , Animales , Axones/metabolismo , Regeneración Nerviosa/genética , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Mitocondrias/metabolismo , Mitocondrias/genética , Ratones , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/patología , Traumatismos del Nervio Óptico/genética , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/genética , Ratones Endogámicos C57BL , Adenosina Trifosfato/metabolismo , Metabolismo Energético/genética
3.
Sci Rep ; 14(1): 23118, 2024 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-39366989

RESUMEN

The retina is part of the central nervous system (CNS). Neurons in the CNS and retinal ganglion cells lack the ability to regenerate axons spontaneously after injury. The intrinsic axonal growth regulators, their interaction and roles that enable or inhibit axon growth are still largely unknown. This study endeavored to characterize the molecular characteristics under neurodegenerative and regenerative conditions. Data-Independent Acquisition Mass Spectrometry was used to map the comprehensive proteome of the regenerative retina from 14-day-old mice (Reg-P14) and adult mice after lens injury (Reg-LI) both showing regrowing axons in vitro, untreated adult mice, and retina from adult mice subjected to two weeks of elevated intraocular pressure showing degeneration. A total of 5750 proteins were identified (false discovery rate < 1%). Proteins identified in both Reg-P14 and Reg-LI groups were correlated to thyroid hormone, Notch, Wnt, and VEGF signaling pathways. Common interactors comprising E1A binding protein P300 (EP300), CREB binding protein (CBP), calcium/calmodulin dependent protein kinase II alpha (CaMKIIα) and sirtuin 1 (SIRT1) were found in both Reg-P14 and Reg-LI retinas. Proteins identified in both regenerating and degenerative groups were correlated to thyroid hormone, Notch, mRNA surveillance and measles signaling pathways, along with PD-L1 expression and the PD-1 checkpoint pathway. Common interactors across regenerative and degenerative retinas comprising NF-kappa-B p65 subunit (RELA), RNA-binding protein with serine-rich domain 1 (RNPS1), EP300 and SIN3 transcription regulator family member A (SIN3A). The findings from our study provide the first mapping of regenerative mechanisms across postnatal, mature and degenerative mouse retinas, revealing potential biomarkers that could facilitate neuro-regeneration in glaucoma.


Asunto(s)
Glaucoma , Regeneración Nerviosa , Proteómica , Retina , Animales , Ratones , Glaucoma/metabolismo , Glaucoma/fisiopatología , Glaucoma/patología , Proteómica/métodos , Regeneración Nerviosa/fisiología , Retina/metabolismo , Biomarcadores/metabolismo , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Proteoma/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
4.
Elife ; 132024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39382568

RESUMEN

Acute retinal ischemia and ischemia-reperfusion injury are the primary causes of retinal neural cell death and vision loss in retinal artery occlusion (RAO). The absence of an accurate mouse model for simulating the retinal ischemic process has hindered progress in developing neuroprotective agents for RAO. We developed a unilateral pterygopalatine ophthalmic artery occlusion (UPOAO) mouse model using silicone wire embolization combined with carotid artery ligation. The survival of retinal ganglion cells and visual function were evaluated to determine the duration of ischemia. Immunofluorescence staining, optical coherence tomography, and haematoxylin and eosin staining were utilized to assess changes in major neural cell classes and retinal structure degeneration at two reperfusion durations. Transcriptomics was employed to investigate alterations in the pathological process of UPOAO following ischemia and reperfusion, highlighting transcriptomic differences between UPOAO and other retinal ischemia-reperfusion models. The UPOAO model successfully replicated the acute interruption of retinal blood supply observed in RAO. 60 min of Ischemia led to significant loss of major retinal neural cells and visual function impairment. Notable thinning of the inner retinal layer, especially the ganglion cell layer, was evident post-UPOAO. Temporal transcriptome analysis revealed various pathophysiological processes related to immune cell migration, oxidative stress, and immune inflammation during the non-reperfusion and reperfusion periods. A pronounced increase in microglia within the retina and peripheral leukocytes accessing the retina was observed during reperfusion periods. Comparison of differentially expressed genes (DEGs) between the UPOAO and high intraocular pressure models revealed specific enrichments in lipid and steroid metabolism-related genes in the UPOAO model. The UPOAO model emerges as a novel tool for screening pathogenic genes and promoting further therapeutic research in RAO.


Asunto(s)
Modelos Animales de Enfermedad , Daño por Reperfusión , Animales , Ratones , Daño por Reperfusión/genética , Oclusión de la Arteria Retiniana/genética , Oclusión de la Arteria Retiniana/etiología , Oclusión de la Arteria Retiniana/patología , Masculino , Ratones Endogámicos C57BL , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Transcriptoma , Retina/patología , Retina/metabolismo , Arteria Retiniana/patología , Isquemia/genética
5.
Invest Ophthalmol Vis Sci ; 65(12): 13, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39382882

RESUMEN

Purpose: The optic nerve head (ONH) is well known to be the initial site of glaucomatous damage; however, the molecular mechanisms initiating this pathology are not fully understood. To further understand the initiating factors in glaucomatous damage we utilized a novel mouse model of glaucoma, B6.EDA+/+ mice, which constitutively express fibronectin containing the extra domain A (FN+EDA). FN+EDA is a known damage-associated molecular pattern (DAMP) that activates Toll-like receptor 4 and elicits a fibro-inflammatory response. Methods: Eyes from B6.EDA+/+ and C57BL/6J mice were evaluated for retinal ganglion cell (RGC) death, retinal nerve fiber layer (RNFL) thickness, and optic nerve (ON) damage at 12 months and 22 months of age. ONH sections were isolated using laser capture microdissection for subsequent RNA-sequencing and Gene Set Enrichment Analysis (GSEA). GSEA results were confirmed using immunohistochemical (IHC) staining. Results: B6.EDA+/+ mice exhibit significantly higher intraocular pressure, loss of RGCs, thinning of the RNFL, and progressive levels of ON damage at 12 months and 22 months of age compared to C57BL/6J controls. Protein expression of DAMPs FN+EDA and biglycan was significantly increased in B6.EDA+/+ mice compared to C57BL/6J controls. GSEA analysis identified significantly up- and downregulated gene groupings at both 12 months and 22 months of age, and IHC staining at 12 and 18 months of age demonstrated significant increases of IFNα, IFNß, and pSTAT1 expression in B6.EDA+/+ mice compared to C57BL/6J controls. Conclusions: Our study characterizes glaucomatous changes to the retina, ON, and ONH over the course of 2 years and identifies novel molecular pathways associated with these pathophysiological changes. These data illustrate the effects of FN+EDA on the fibro-inflammatory response in the aging ONH in a novel mouse model of glaucoma.


Asunto(s)
Modelos Animales de Enfermedad , Fibronectinas , Glaucoma , Presión Intraocular , Ratones Endogámicos C57BL , Disco Óptico , Células Ganglionares de la Retina , Animales , Ratones , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Disco Óptico/patología , Disco Óptico/metabolismo , Presión Intraocular/fisiología , Glaucoma/metabolismo , Glaucoma/genética , Fibronectinas/metabolismo , Fibronectinas/genética , Fibras Nerviosas/patología , Masculino , Enfermedades del Nervio Óptico/genética , Enfermedades del Nervio Óptico/metabolismo , Tomografía de Coherencia Óptica
6.
Acta Neuropathol Commun ; 12(1): 149, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39267142

RESUMEN

Motor neuron loss is well recognized in amyotrophic lateral sclerosis (ALS), but research on retinal ganglion cells (RGCs) is limited. Ocular symptoms are generally not considered classic ALS symptoms, although RGCs and spinal motor neurons share certain cell pathologies, including hallmark signs of glutamate neurotoxicity, which may be triggered by activation of extrasynaptic NMDA receptors (NMDARs). To explore potential novel strategies to prevent ALS-associated death of RGCs, we utilized inhibition of the TwinF interface, a new pharmacological principle that detoxifies extrasynaptic NMDARs by disrupting the NMDAR/TRPM4 death signaling complex. Using the ALS mouse model SOD1G93A, we found that the small molecule TwinF interface inhibitor FP802 prevents the loss of RGCs, improves pattern electroretinogram (pERG) performance, increases the retinal expression of Bdnf, and restores the retinal expression of the immediate early genes, Inhibin beta A and Npas4. Thus, FP802 not only prevents, as recently described, death of spinal motor neurons in SOD1G93A mice, but it also mitigates ALS-associated retinal damage. TwinF interface inhibitors have great potential for alleviating neuro-ophthalmologic symptoms in ALS patients and offer a promising new avenue for therapeutic intervention.


Asunto(s)
Esclerosis Amiotrófica Lateral , Modelos Animales de Enfermedad , Ratones Transgénicos , Células Ganglionares de la Retina , Animales , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Ratones , Electrorretinografía , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Humanos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo
7.
Neuromolecular Med ; 26(1): 37, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39266914

RESUMEN

As the primary connection between the eye and brain, the optic nerve plays a pivotal role in visual information transmission. Injuries to the optic nerve can occur for various reasons, including trauma, glaucoma, and neurodegenerative diseases. Retinal ganglion cells (RGCs), a type of neurons that extend axons through the optic nerve, can rapidly respond to injury and initiate cell death. Additionally, following optic nerve injury microglia, which serve as markers of neuroinflammation, transition from a resting state to an activated state. The phosphorylation of collapsin response mediator protein2 (CRMP2) in the semaphorin 3A (Sema3A) signalling pathway affects several processes, including axon guidance and neuron regeneration. In this study, we used an optic nerve crush (ONC) mouse model to investigate the effects of suppressing CRMP2 phosphorylation on microglia activation. We found that CRMP2 phosphorylation inhibitor suppressed RGCs loss and promoted neuronal regeneration following ONC. In addition, CRMP2 S522A mutant (CRMP2 KI) mice exhibited decreased microglial activation in both the retina and optic nerve following ONC. These results suggest that inhibiting the phosphorylation of CRMP2 can alleviate the loss of RGCs and microglial activation after optic nerve injury, providing insight into the development of treatments for optical neuropathies and neurodegenerative diseases.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular , Microglía , Regeneración Nerviosa , Proteínas del Tejido Nervioso , Traumatismos del Nervio Óptico , Nervio Óptico , Retina , Células Ganglionares de la Retina , Semaforina-3A , Animales , Traumatismos del Nervio Óptico/fisiopatología , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/tratamiento farmacológico , Microglía/metabolismo , Microglía/efectos de los fármacos , Fosforilación , Ratones , Regeneración Nerviosa/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas del Tejido Nervioso/genética , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Nervio Óptico/metabolismo , Semaforina-3A/genética , Semaforina-3A/metabolismo , Retina/efectos de los fármacos , Retina/metabolismo , Compresión Nerviosa , Ratones Endogámicos C57BL , Masculino , Modelos Animales de Enfermedad , Ratones Transgénicos
8.
J Comp Neurol ; 532(8): e25663, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39235164

RESUMEN

The transcription factor forkhead/winged-helix domain proteins Foxp1 and Foxp2 have previously been studied in mouse retina, where they are expressed in retinal ganglion cells named F-mini and F-midi. Here we show that both transcription factors are expressed by small subpopulations (on average less than 10%) of retinal ganglion cells in the retina of the marmoset monkey (Callithrix jacchus). The morphology of Foxp1- and Foxp2-expressing cells was revealed by intracellular DiI injections of immunofluorescent cells. Foxp1- and Foxp2-expressing cells comprised multiple types of wide-field ganglion cells, including broad thorny cells, narrow thorny cells, and tufted cells. The large majority of Foxp2-expressing cells were identified as tufted cells. Tufted cells stratify broadly in the middle of the inner plexiform layer. They resemble broad thorny cells but their proximal dendrites are bare of branches and the distal dendrites branch frequently forming dense dendritic tufts. Double labeling with calretinin, a previously established marker for broad thorny and narrow thorny cells, showed that only a small proportion of ganglion cells co-expressed calretinin and Foxp1 or Foxp2 supporting the idea that the two markers are differentially expressed in retinal ganglion cells of marmoset retina.


Asunto(s)
Callithrix , Factores de Transcripción Forkhead , Células Ganglionares de la Retina , Animales , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/biosíntesis , Células Ganglionares de la Retina/metabolismo , Masculino , Femenino , Retina/metabolismo , Retina/citología
9.
Invest Ophthalmol Vis Sci ; 65(11): 8, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39230992

RESUMEN

Purpose: This study investigates alterations in intrinsically photosensitive retinal ganglion cells (ipRGCs) and dopaminergic amacrine cells (DACs) in lid suture myopia (LSM) rats. Methods: LSM was induced in rats by suturing the right eyes for 4 weeks. Double immunofluorescence staining of ipRGCs and DACs in whole-mount retinas was performed to analyze changes in the density and morphology of control, LSM, and fellow eyes. Real-time quantitative PCR and Western blotting were used to detect related genes and protein expression levels. Results: Significant myopia was induced in the lid-sutured eye, but the fellow eye was not different to control. Decreased ipRGC density with paradoxically increased overall melanopsin expression and enlarged dendritic beads was observed in both the LSM and fellow eyes of the LSM rat retinas. In contrast, DAC changes occurred only in the LSM eyes, with reduced DAC density and tyrosine hydroxylase (TH) expression, sparser dendritic processes, and fewer varicosities. Interestingly, contacts between ipRGCs and DACs in the inner plexiform layer (IPL) and the expression of pituitary adenylate cyclase-activating polypeptide (PACAP) and vesicular monoamine transporter protein 2 (VMAT2) mRNA were decreased in the LSM eyes. Conclusions: The ipRGCs and DACs in LSM rat retinas undergo multiple alterations in density, morphology, and related molecule expressions. However, the ipRGC changes alone appear not to be required for the development of myopia, given that myopia is only induced in the lid-sutured eye, and they are unlikely alone to drive the DAC changes. Reduced contacts between ipRGCs and DACs in the LSM eyes may be the structural foundation for the impaired signaling between them. PACAP and VMAT2, strongly associated with ipRGCs and DACs, may play important roles in LSM through complex mechanisms.


Asunto(s)
Células Amacrinas , Western Blotting , Modelos Animales de Enfermedad , Miopía , Células Ganglionares de la Retina , Opsinas de Bastones , Animales , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Ratas , Miopía/metabolismo , Células Amacrinas/metabolismo , Células Amacrinas/patología , Opsinas de Bastones/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/genética , Masculino , Ratas Sprague-Dawley , Párpados/patología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Recuento de Células , Proteína 2 de Transporte Vesicular de Glutamato
10.
Transl Vis Sci Technol ; 13(9): 27, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39330985

RESUMEN

Purpose: Resveratrol (RSV) is a nutraceutical compound known for its therapeutic potential in neurodegenerative and metabolic diseases. RSV promotes survival signals in retinal ganglion cells (RGCs) through activation of SIRT1, an NAD+-dependent deacetylase. RSV and SIRT1 reduce RGC loss induced by direct optic nerve injury, but effects in indirect models of traumatic optic neuropathy remain unknown and are examined in this study. Methods: An electromagnetic stereotaxic impactor device was used to impart five traumatic skull impacts with an inter-concussion interval of 48 hours to wild type (WT) and SIRT1 knock in (KI) C57BL/6J mice overexpressing the SIRT1 gene. A cohort of WT mice also received intranasal administration of RSV (16 mg/kg) throughout the experimental period. Loss of righting reflex (RR), optokinetic response (OKR) scores, and immunolabeled RGC count are determined to assess optic neuropathy in this model of traumatic brain injury (TBI). Results: TBI significantly decreases RGC survival and decreases OKR scores compared with control uninjured mice. Either RSV administration in WT mice, or SIRT1 overexpression in SIRT1 KI mice, significantly increases RGC survival and improves OKR scores. RR time increases after the first few impacts in all groups of mice subjected to TBI, demonstrating that RSV and SIRT1 overexpression are able to attenuate optic neuropathy following similar degrees of TBI. Conclusions: Intranasal RSV is effective in preserving visual function in WT mice following TBI. Constitutive overexpression of SIRT1 recapitulates the neuroprotective effect of RSV. Translational Relevance: Results support future exploration of RSV as a potential therapy for indirect traumatic optic neuropathy.


Asunto(s)
Modelos Animales de Enfermedad , Traumatismos Cerrados de la Cabeza , Ratones Endogámicos C57BL , Traumatismos del Nervio Óptico , Resveratrol , Células Ganglionares de la Retina , Sirtuina 1 , Animales , Sirtuina 1/genética , Sirtuina 1/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Traumatismos del Nervio Óptico/tratamiento farmacológico , Traumatismos del Nervio Óptico/genética , Traumatismos del Nervio Óptico/patología , Ratones , Resveratrol/farmacología , Resveratrol/uso terapéutico , Resveratrol/administración & dosificación , Traumatismos Cerrados de la Cabeza/genética , Traumatismos Cerrados de la Cabeza/patología , Traumatismos Cerrados de la Cabeza/tratamiento farmacológico , Masculino , Administración Intranasal , Supervivencia Celular/efectos de los fármacos , Ratones Transgénicos , Reflejo de Enderezamiento/efectos de los fármacos , Nistagmo Optoquinético/efectos de los fármacos
11.
Biomolecules ; 14(9)2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39334813

RESUMEN

Rotenone is a mitochondrial complex I inhibitor that causes retinal degeneration. A study of a rat model of rotenone-induced retinal degeneration suggested that this model is caused by indirect postsynaptic N-methyl-D-aspartate (NMDA) stimulation triggered by oxidative stress-mediated presynaptic intracellular calcium signaling. To elucidate the mechanisms by which rotenone causes axonal degeneration, we investigated morphological changes in optic nerves and the change in retinal ganglion cell (RGC) number in rats. Optic nerves and retinas were collected 3 and 7 days after the intravitreal injection of rotenone. The cross-sections of the optic nerves were subjected to a morphological analysis with axon quantification. The axons and somas of RGCs were analyzed immunohistochemically in retinal flatmounts. In the optic nerve, rotenone induced axonal swelling and degeneration with the incidence of reactive gliosis. Rotenone also significantly reduced axon numbers in the optic nerve. Furthermore, rotenone caused axonal thinning, fragmentation, and beading in RGCs on flatmounts and decreased the number of RGC soma. In conclusion, the intravitreal injection of rotenone in rats induced morphological abnormities with a reduced number of optic nerve axons and RGC axons when the RGC somas were degenerated. These findings help elucidate the pathogenesis of optic neuropathy induced by mitochondrial dysfunction.


Asunto(s)
Axones , Traumatismos del Nervio Óptico , Células Ganglionares de la Retina , Rotenona , Animales , Rotenona/toxicidad , Rotenona/efectos adversos , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Ratas , Masculino , Axones/patología , Axones/efectos de los fármacos , Axones/metabolismo , Traumatismos del Nervio Óptico/patología , Traumatismos del Nervio Óptico/inducido químicamente , Traumatismos del Nervio Óptico/metabolismo , Nervio Óptico/patología , Nervio Óptico/efectos de los fármacos , Nervio Óptico/metabolismo , Ratas Sprague-Dawley , Inyecciones Intravítreas
12.
PLoS One ; 19(9): e0309400, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39255314

RESUMEN

Glaucoma is a blinding disease where the retinal ganglion cells and their axons degenerate. Degradation of axonal microtubules is thought to play a critical role in the pathogenesis, but the mechanism is unknown. Here we investigate whether microtubule disruption in glaucoma can be alleviated by metabolic rescue. The integrity of axonal microtubules and the morphology of the retinal nerve fibers were evaluated by second-harmonic generation microscopy in a mouse model of glaucoma, DBA/2J, which received a dietary supplement of nicotinamide (NAM) for reducing metabolic stress. It was compared with control DBA/2J, which did not receive NAM, and non-glaucomatous DBA/2J-Gpnmb+. We found that the morphology of the retinal nerve fibers, but not axonal microtubules, are significantly protected by NAM. The decoupling is analogous to microtubule deficit, a glaucoma pathology in which axonal microtubules exhibit rapid degradation compared to the morphology of the retinal nerve fibers. Understanding microtubule deficit could provide insights into the divergent responses to NAM. From co-registered images of second-harmonic generation and immunofluorescence, it was determined that microtubule deficit was not due to a shortage of tubulins. Furthermore, microtubule deficit colocalized with the sectors in which the retinal ganglion cells were disconnected from the brain, suggesting that microtubule disruption is associated with axonal transport deficit in glaucoma. Together, our data suggests significant role axonal microtubules play in glaucomatous degeneration, offering a new opportunity for neuroprotection.


Asunto(s)
Modelos Animales de Enfermedad , Glaucoma , Ratones Endogámicos DBA , Microtúbulos , Niacinamida , Células Ganglionares de la Retina , Animales , Glaucoma/patología , Glaucoma/metabolismo , Glaucoma/tratamiento farmacológico , Niacinamida/farmacología , Niacinamida/uso terapéutico , Ratones , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Axones/efectos de los fármacos , Axones/metabolismo , Axones/patología , Microscopía/métodos , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/patología , Fibras Nerviosas/metabolismo
13.
J Proteome Res ; 23(10): 4674-4683, 2024 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-39319515

RESUMEN

Metabolic dysfunction plays a crucial role in the pathogenesis of glaucoma. In this study, we used Olink proteomics profiling to identify potential biomarkers for glaucoma. Aqueous humor samples were obtained from 44 cataract patients and 44 glaucoma patients. We identified 84 differentially expressed metabolic proteins between the glaucoma and the cataract group. Gene Ontology enrichment analysis highlighted the involvement of these proteins in ER-associated degradation pathway, regulation of interleukin-13 production, and DNA damage response pathway. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis further revealed links to pathways, such as tyrosine and pyrimidine metabolism. Among these, ALDH1A1 emerged as a candidate with a significant diagnostic potential for glaucoma. ALDH1A1 also exhibited a prominent role in the protein-protein interaction network. Elevated levels of ALDH1A1 in the aqueous humor of glaucoma patients were confirmed both in clinical samples and in an ischemia/reperfusion model. Functional assays confirmed that elevated ALDH1A1 induced retinal ganglion cell (RGC) apoptosis in vitro and demonstrated its pro-apoptotic role in RGCs in vivo. Collectively, these findings not only underscore the significance of ALDH1A1 in glaucoma but also provide valuable insights into clinical decision-making and therapeutic strategies.


Asunto(s)
Familia de Aldehído Deshidrogenasa 1 , Humor Acuoso , Biomarcadores , Glaucoma , Proteómica , Humanos , Glaucoma/metabolismo , Glaucoma/genética , Glaucoma/patología , Biomarcadores/metabolismo , Proteómica/métodos , Humor Acuoso/metabolismo , Familia de Aldehído Deshidrogenasa 1/metabolismo , Familia de Aldehído Deshidrogenasa 1/genética , Retinal-Deshidrogenasa/metabolismo , Retinal-Deshidrogenasa/genética , Femenino , Masculino , Mapas de Interacción de Proteínas , Apoptosis/genética , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Anciano , Persona de Mediana Edad , Animales , Catarata/metabolismo , Catarata/genética
14.
Acta Neuropathol Commun ; 12(1): 150, 2024 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-39300576

RESUMEN

BACKGROUND: Glaucoma is a leading cause of blindness, affecting retinal ganglion cells (RGCs) and their axons. By 2040, it is likely to affect 110 million people. Neuroinflammation, specifically through the release of proinflammatory cytokines by M1 microglial cells, plays a crucial role in glaucoma progression. Indeed, in post-mortem human studies, pre-clinical models, and ex-vivo models, RGC degeneration has been consistently shown to be linked to inflammation in response to cell death and tissue damage. Recently, Rho kinase inhibitors (ROCKis) have emerged as potential therapies for neuroinflammatory and neurodegenerative diseases. This study aimed to investigate the potential effects of three ROCKis (Y-27632, Y-33075, and H-1152) on retinal ganglion cell (RGC) loss and retinal neuroinflammation using an ex-vivo retinal explant model. METHODS: Rat retinal explants underwent optic nerve axotomy and were treated with Y-27632, Y-33075, or H-1152. The neuroprotective effects on RGCs were evaluated using immunofluorescence and Brn3a-specific markers. Reactive glia and microglial activation were studied by GFAP, CD68, and Iba1 staining. Flow cytometry was used to quantify day ex-vivo 4 (DEV 4) microglial proliferation and M1 activation by measuring the number of CD11b+, CD68+, and CD11b+/CD68+ cells after treatment with control solvent or Y-33075. The modulation of gene expression was measured by RNA-seq analysis on control and Y-33075-treated explants and glial and pro-inflammatory cytokine gene expression was validated by RT-qPCR. RESULTS: Y-27632 and H-1152 did not significantly protect RGCs. By contrast, at DEV 4, 50 µM Y-33075 significantly increased RGC survival. Immunohistology showed a reduced number of Iba1+/CD68+ cells and limited astrogliosis with Y-33075 treatment. Flow cytometry confirmed lower CD11b+, CD68+, and CD11b+/CD68+ cell numbers in the Y-33075 group. RNA-seq showed Y-33075 inhibited the expression of M1 microglial markers (Tnfα, Il-1ß, Nos2) and glial markers (Gfap, Itgam, Cd68) and to reduce apoptosis, ferroptosis, inflammasome formation, complement activation, TLR pathway activation, and P2rx7 and Gpr84 gene expression. Conversely, Y-33075 upregulated RGC-specific markers, neurofilament formation, and neurotransmitter regulator expression, consistent with its neuroprotective effects. CONCLUSION: Y-33075 demonstrates marked neuroprotective and anti-inflammatory effects, surpassing the other tested ROCKis (Y-27632 and H-1152) in preventing RGC death and reducing microglial inflammatory responses. These findings highlight its potential as a therapeutic option for glaucoma.


Asunto(s)
Fármacos Neuroprotectores , Piridinas , Células Ganglionares de la Retina , Quinasas Asociadas a rho , Animales , Piridinas/farmacología , Quinasas Asociadas a rho/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Fármacos Neuroprotectores/farmacología , Ratas , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Retina/efectos de los fármacos , Retina/patología , Retina/metabolismo , Amidas/farmacología , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Ratas Sprague-Dawley , Neuroprotección/efectos de los fármacos , Neuroprotección/fisiología , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Inhibidores de Proteínas Quinasas/farmacología , Masculino , Traumatismos del Nervio Óptico/tratamiento farmacológico , Traumatismos del Nervio Óptico/patología , Traumatismos del Nervio Óptico/metabolismo , Isoquinolinas , Sulfonamidas
15.
Acta Neuropathol Commun ; 12(1): 137, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39180087

RESUMEN

A compromised capacity to maintain NAD pools is recognized as a key underlying pathophysiological feature of neurodegenerative diseases. NAD acts as a substrate in major cell functions including mitochondrial homeostasis, cell signalling, axonal transport, axon/Wallerian degeneration, and neuronal energy supply. Dendritic degeneration is an early marker of neuronal stress and precedes cell loss. However, little is known about dendritic structural preservation in pathologic environments and remodelling in mature neurons. Retinal ganglion cell dendritic atrophy is an early pathological feature in animal models of the disease and has been demonstrated in port-mortem human glaucoma samples. Here we report that a nicotinamide (a precursor to NAD through the NAD salvage pathway) enriched diet provides robust retinal ganglion cell dendritic protection and preserves dendritic structure in a rat model of experimental glaucoma. Metabolomic analysis of optic nerve samples from the same animals demonstrates that nicotinamide provides robust metabolic neuroprotection in glaucoma. Advances in our understanding of retinal ganglion cell metabolic profiles shed light on the energetic shift that triggers early neuronal changes in neurodegenerative diseases. As nicotinamide can improve visual function short term in existing glaucoma patients, we hypothesize that a portion of this visual recovery may be due to dendritic preservation in stressed, but not yet fully degenerated, retinal ganglion cells.


Asunto(s)
Modelos Animales de Enfermedad , Glaucoma , Fármacos Neuroprotectores , Niacinamida , Células Ganglionares de la Retina , Animales , Niacinamida/farmacología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Glaucoma/metabolismo , Glaucoma/patología , Fármacos Neuroprotectores/farmacología , Ratas , Relación Dosis-Respuesta a Droga , Masculino , Administración Oral , Nervio Óptico/efectos de los fármacos , Nervio Óptico/patología , Nervio Óptico/metabolismo , Neuroprotección/efectos de los fármacos , Neuroprotección/fisiología , Dendritas/efectos de los fármacos , Dendritas/patología , Dendritas/metabolismo , Complejo Vitamínico B/farmacología , Complejo Vitamínico B/administración & dosificación
16.
Biomed Pharmacother ; 178: 117270, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39126773

RESUMEN

The blood supply in the retina ensures photoreceptor function and maintains regular vision. Leber's hereditary optic neuropathy (LHON), caused by the mitochondrial DNA mutations that deteriorate complex I activity, is characterized by progressive vision loss. Although some reports indicated retinal vasculature abnormalities as one of the comorbidities in LHON, the paracrine influence of LHON-affected retinal ganglion cells (RGCs) on vascular endothelial cell physiology remains unclear. To address this, we established an in vitro model of mitochondrial complex I deficiency using induced pluripotent stem cell-derived RGCs (iPSC-RGCs) treated with a mitochondrial complex I inhibitor rotenone (Rot) to recapitulate LHON pathologies. The secretomes from Rot-treated iPSC-RGCs (Rot-iPSC-RGCs) were collected, and their treatment effect on human umbilical vein endothelial cells (HUVECs) was studied. Rot induced LHON-like characteristics in iPSC-RGCs, including decreased mitochondrial complex I activity and membrane potential, and increased mitochondrial reactive oxygen species (ROS) and apoptosis, leading to mitochondrial dysfunction. When HUVECs were exposed to conditioned media (CM) from Rot-iPSC-RGCs, the angiogenesis of HUVECs was suppressed compared to those treated with CM from control iPSC-RGCs (Ctrl-iPSC-RGCs). Angiogenesis-related proteins were altered in the secretomes from Rot-iPSC-RGC-derived CM, particularly angiopoietin, MMP-9, uPA, collagen XVIII, and VEGF were reduced. Notably, GeneMANIA analysis indicated that VEGFA emerged as the pivotal angiogenesis-related protein among the identified proteins secreted by health iPSC-RGCs but reduced in the secretomes from Rot-iPSC-RGCs. Quantitative real-time PCR and western blots confirmed the reduction of VEGFA at both transcription and translation levels, respectively. Our study reveals that Rot-iPSC-RGCs establish a microenvironment to diminish the angiogenic potential of vascular cells nearby, shedding light on the paracrine regulation of LHON-affected RGCs on retinal vasculature.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana , Células Madre Pluripotentes Inducidas , Atrofia Óptica Hereditaria de Leber , Células Ganglionares de la Retina , Humanos , Atrofia Óptica Hereditaria de Leber/metabolismo , Atrofia Óptica Hereditaria de Leber/patología , Atrofia Óptica Hereditaria de Leber/genética , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Rotenona/farmacología , Medios de Cultivo Condicionados/farmacología , Apoptosis/efectos de los fármacos , Complejo I de Transporte de Electrón/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Neovascularización Patológica/metabolismo , Angiogénesis
17.
Mol Brain ; 17(1): 58, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39175067

RESUMEN

Protocadherin 19 (Pcdh19) is a homophilic cell adhesion molecule and is involved in a variety of neuronal functions. Here, we tested whether Pcdh19 has a regulatory role in axon guidance using the developing Xenopus retinotectal system. We performed targeted microinjections of a translation blocking antisense morpholino oligonucleotide to knock down the expression of Pcdh19 selectively in the central nervous system. Knocking down Pcdh19 expression resulted in navigational errors of retinal ganglion cell (RGC) axons specifically at the optic chiasm. Instead of projecting to the contralateral optic tectum, RGC axons in the Pcdh19-depleted embryo misprojected ipsilaterally. Although incorrectly delivered into the ipsilateral brain hemisphere, these axons correctly reached the optic tectum. These data suggest that Pcdh19 has a critical role in preventing mixing of RGC axons originating from the opposite eyes at the optic chiasm, highlighting the importance of cell adhesion in bundling of RGC axons.


Asunto(s)
Orientación del Axón , Axones , Cadherinas , Protocadherinas , Células Ganglionares de la Retina , Proteínas de Xenopus , Xenopus laevis , Animales , Cadherinas/metabolismo , Proteínas de Xenopus/metabolismo , Proteínas de Xenopus/genética , Células Ganglionares de la Retina/metabolismo , Xenopus laevis/embriología , Axones/metabolismo , Retina/metabolismo , Retina/embriología , Vías Visuales , Técnicas de Silenciamiento del Gen , Quiasma Óptico/embriología , Quiasma Óptico/metabolismo , Colículos Superiores/embriología , Colículos Superiores/metabolismo , Regulación del Desarrollo de la Expresión Génica
18.
Invest Ophthalmol Vis Sci ; 65(10): 34, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39186263

RESUMEN

Purpose: In response to hypoxia, sympathetic fibers to the retina activate ß-adrenoceptors (ß-ARs) that play an important role in the regulation of vascular and neuronal functions. We investigated the role of ß3-AR using the mouse model of oxygen-induced retinopathy (OIR). Methods: Mouse pups were exposed to 75% oxygen at postnatal day 7 (PD7) followed by a return to room air at PD12. The ß3-AR preferential agonist BRL37344 was subcutaneously administered once daily at different times after the return to room air. At PD17, the OIR mice underwent flash and pattern electroretinogram. After sacrifice, retinal wholemounts were used for vessel staining or immunohistochemistry for astrocytes, Müller cells, or retinal ganglion cells (RGCs). In retinal homogenates, the levels of markers associated with neovascularization (NV), the blood-retinal barrier (BRB), or astrocytes were determined by western blot, and quantitative reverse-transcription polymerase chain reaction was used to assess ß3-AR messenger. Administration of the ß3-AR antagonist SR59230A was performed to verify BRL37344 selectivity. Results: ß3-AR expression is upregulated in response to hypoxia, but its increase is prevented by BRL37344, which counteracts NV by inhibiting the pro-angiogenic pathway, activating the anti-angiogenic pathway, recovering BRB-associated markers, triggering nitric oxide production, and favoring revascularization of the central retina through recovered density of astrocytes that ultimately counteracts NV in the midperiphery. Vasculature rescue prevents dysfunctional retinal activity and counteracts OIR-associated retinal ganglion cell loss. Conclusions: ß3-AR has emerged as a crucial intermediary in hypoxia-dependent NV, suggesting a role of ß3-AR agonists in the treatment of proliferative retinopathies.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 3 , Modelos Animales de Enfermedad , Electrorretinografía , Ratones Endogámicos C57BL , Oxígeno , Receptores Adrenérgicos beta 3 , Neovascularización Retiniana , Animales , Ratones , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/prevención & control , Neovascularización Retiniana/patología , Oxígeno/toxicidad , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Receptores Adrenérgicos beta 3/metabolismo , Animales Recién Nacidos , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Etanolaminas/farmacología , Vasos Retinianos/efectos de los fármacos , Retinopatía de la Prematuridad/metabolismo , Retinopatía de la Prematuridad/tratamiento farmacológico , Astrocitos/metabolismo , Astrocitos/efectos de los fármacos , Inmunohistoquímica , Angiogénesis
19.
Sci Adv ; 10(35): eadk4062, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39196935

RESUMEN

The mammalian retina is considered an autonomous circuit, yet work dating back to Ramon y Cajal indicates that it receives inputs from the brain. How such inputs affect retinal processing has remained unknown. We confirmed brain-to-retina projections of histaminergic neurons from the mouse hypothalamus. Histamine application ex vivo altered the activity of various retinal ganglion cells (RGCs), including direction-selective RGCs that gained responses to high motion velocities. These results were reproduced in vivo with optic tract recordings where histaminergic retinopetal axons were activated chemogenetically. Such changes could improve vision of fast-moving objects (e.g., while running), which fits with the known increased activity of histaminergic neurons during arousal. An antihistamine drug reduced optomotor responses to high-speed moving stimuli in freely moving mice. In humans, the same antihistamine nonuniformly modulated visual sensitivity across the visual field, indicating an evolutionary conserved function of the histaminergic system. Our findings expose a previously unappreciated role for brain-to-retina projections in modulating retinal function.


Asunto(s)
Histamina , Hipotálamo , Retina , Células Ganglionares de la Retina , Animales , Histamina/farmacología , Histamina/metabolismo , Hipotálamo/metabolismo , Hipotálamo/citología , Hipotálamo/fisiología , Ratones , Retina/metabolismo , Retina/fisiología , Retina/efectos de los fármacos , Retina/citología , Células Ganglionares de la Retina/fisiología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Neuronas/efectos de los fármacos , Humanos , Ratones Endogámicos C57BL , Vías Visuales/efectos de los fármacos , Vías Visuales/fisiología
20.
Development ; 151(15)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39133185

RESUMEN

Retinal regeneration has been mostly studied after widespread tissue injury, but it is not well understood how the retina regenerates at the cellular level following loss of specific cell types. In a new study, Jeff Mumm and colleagues selectively ablate retinal ganglion cells in zebrafish and find that the retina elicits different genetic responses in a context-dependent manner to replace lost cells. To find out more about the story behind the paper, we caught up with first author Kevin Emmerich and corresponding author Jeff Mumm, Associate Professor in Ophthalmology at Johns Hopkins University.


Asunto(s)
Pez Cebra , Animales , Humanos , Historia del Siglo XXI , Células Ganglionares de la Retina/fisiología , Células Ganglionares de la Retina/metabolismo , Retina , Historia del Siglo XX , Regeneración/fisiología , Oftalmología/historia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA