Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Nature ; 626(7999): 574-582, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38086421

ABSTRACT

The intrinsic mechanisms that regulate neurotoxic versus neuroprotective astrocyte phenotypes and their effects on central nervous system degeneration and repair remain poorly understood. Here we show that injured white matter astrocytes differentiate into two distinct C3-positive and C3-negative reactive populations, previously simplified as neurotoxic (A1) and neuroprotective (A2)1,2, which can be further subdivided into unique subpopulations defined by proliferation and differential gene expression signatures. We find the balance of neurotoxic versus neuroprotective astrocytes is regulated by discrete pools of compartmented cyclic adenosine monophosphate derived from soluble adenylyl cyclase and show that proliferating neuroprotective astrocytes inhibit microglial activation and downstream neurotoxic astrocyte differentiation to promote retinal ganglion cell survival. Finally, we report a new, therapeutically tractable viral vector to specifically target optic nerve head astrocytes and show that raising nuclear or depleting cytoplasmic cyclic AMP in reactive astrocytes inhibits deleterious microglial or macrophage cell activation and promotes retinal ganglion cell survival after optic nerve injury. Thus, soluble adenylyl cyclase and compartmented, nuclear- and cytoplasmic-localized cyclic adenosine monophosphate in reactive astrocytes act as a molecular switch for neuroprotective astrocyte reactivity that can be targeted to inhibit microglial activation and neurotoxic astrocyte differentiation to therapeutic effect. These data expand on and define new reactive astrocyte subtypes and represent a step towards the development of gliotherapeutics for the treatment of glaucoma and other optic neuropathies.


Subject(s)
Astrocytes , Neuroprotection , Adenylyl Cyclases/metabolism , Astrocytes/cytology , Astrocytes/enzymology , Astrocytes/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Cell Survival , Cyclic AMP/metabolism , Cytoplasm/metabolism , Macrophages/metabolism , Macrophages/pathology , Microglia/metabolism , Microglia/pathology , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Optic Nerve Injuries/therapy , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , White Matter/metabolism , White Matter/pathology , Glaucoma/pathology , Glaucoma/therapy
2.
Ann Diagn Pathol ; 61: 152059, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36343605

ABSTRACT

OBJECTIVE: The aim was to evaluate the main indications for prenatal diagnosis, the prevalence of abnormal copy number variations (CNVs), correlate them with clinical findings, analyze the prevalence of VUS, report the rare variants found and additionally highlight the clinical importance of microarray-based comparative genomic hybridization (aCGH) in prenatal diagnosis. STUDY DESIGN: We retrospectively analyzed a cohort of 772 fetuses with indication for genetic study in two tertiary hospitals, in a 9-years-period, using aCGH. RESULTS: Our results demonstrated 8.3 % (6.4-10.5 %, 95 % CI) detection rate of pathogenic CNVs. Within this group, the main indication was structural malformations (57 %) mainly involving central nervous system, skeletal and cardiac systems. Pathogenic results in cases with multiple malformations were higher than in cases with isolated anatomical system malformations showing statistical significant differences (p < 0.001). The second indication where we found more pathogenic CNVs was increased nuchal translucency (5-6.4 mm). In fact, the rate of pathogenic CNVs did not show significant differences between structural and non-structural malformations (p > 0.001), highlighting the relevance of genetic study by aCGH also in cases with no structural malformations. A total of 217 fetuses with CNVs classified as VUS were identified, mainly involving chromosomes X, 1 and 16. CONCLUSION: Our findings demonstrate 4.9 % (4.2-5.6 %, 95 % CI) increased in the diagnostic yield using aCGH compared to the use of conventional karyotype alone, confirming that the aCGH can improve the accuracy of prenatal diagnosis. Our survey provides a full genotype-phenotype analysis that can be clinically useful for the classification of variants in the context of prenatal setting, helping to provide a better reproductive genetic counselling.


Subject(s)
Chromosome Aberrations , DNA Copy Number Variations , Pregnancy , Female , Humans , Comparative Genomic Hybridization/methods , Retrospective Studies , Prenatal Diagnosis/methods , Fetus/abnormalities , Genetic Association Studies
3.
Rev. enferm. UERJ ; 29: e62723, jan.-dez. 2021. tab, graf
Article in Portuguese | LILACS-Express | LILACS, BDENF - Nursing | ID: biblio-1365799

ABSTRACT

RESUMO Objetivo identificar atitudes das crianças em relação aos idosos. Método foi realizado um estudo quantitativo, descritivo, exploratório e correlacional. Resultados a população de crianças entre os 3 e os 10 anos. A recolha de dados com alguns dados sociodemográficos da escala "Galvão" é aplicada na população portuguesa. Amostra com 109 crianças entre os 3 e os 10 anos, nacionalidade portuguesa e residentes em Portugal. Os resultados mostraram que em média as crianças (24%) apresentam atitude positiva em relação aos idosos. Este valor esteve próximo do ponto médio da escala (28%), que os coloca numa atitude moderada. Conclusão as crianças apresentaram atitudes mais positivas para com os idosos, embora com alguma inconsistência em algumas dimensões. Pode tornar-se uma contribuição importante para a análise de estereótipos sobre o envelhecimento. É importante ser capaz de intervir nas atitudes das crianças em relação aos idosos, ajudando as crianças a compreender as mudanças normais de envelhecimento.


RESUMEN Objetivo identificar actitudes de niños hacia los ancianos. Método se realizó un estudio cuantitativo, descriptivo, exploratorio y correlacional. Resultados la población estudiada fue de niños entre 3 y 10 años. La recolección con algunos datos sociodemográficos, aplicación de la escala "Galvão" se aplicó en la población portuguesa. La muestra era compuesta por 109 niños entre 3 y 10 años, de nacionalidad portuguesa y residentes en Portugal. Los resultados han demostrado que en promedio los niños (24%) presentan una actitud positiva respecto a los ancianos. Este valor estuvo cerca del punto medio de la escala (28%), lo que los pone en una actitud moderada. Conclusión los niños presentaron actitudes más positivas hacia los ancianos, aunque con cierta inconsistencia en algunas dimensiones. Esta investigación puede convertirse en una importante contribución para analizar los estereotipos sobre el envejecimiento. Es importante ser capaz de intervenir en las actitudes de los niños, ayudándolos a comprender los cambios normales del envejecimiento.


ABSTRACT Objective to identify children's attitudes towards the elderly. Methods a quantitative, descriptive, exploratory, correlational study was conducted. Results the study population was children from 3 to 10 years old. Sociodemographic data were collected and a Galvão scale, validated for the population of Portugal, was applied. The convenience sample comprised 109 children from 3 to 10 years old, of Portuguese nationality and resident in Portugal. On average, the children were found to have a 24% positive attitude towards the elderly. This value was close to the midpoint of the scale (28%), rating their attitude as moderate. Conclusion the children's attitudes towards the elderly were predominantly positive, although with inconsistency in some dimensions. This study may become an important contribution to the analysis of stereotypes about aging. It is important to be able to influence children's attitudes towards the elderly and help them understand the normal changes of aging.

4.
eNeuro ; 8(1)2021.
Article in English | MEDLINE | ID: mdl-33441400

ABSTRACT

The failure of adult CNS neurons to survive and regenerate their axons after injury or in neurodegenerative disease remains a major target for basic and clinical neuroscience. Recent data demonstrated in the adult mouse that exogenous expression of Sry-related high-mobility-box 11 (Sox11) promotes optic nerve regeneration after optic nerve injury but exacerbates the death of a subset of retinal ganglion cells (RGCs), α-RGCs. During development, Sox11 is required for RGC differentiation from retinal progenitor cells (RPCs), and we found that mutation of a single residue to prevent SUMOylation at lysine 91 (K91) increased Sox11 nuclear localization and RGC differentiation in vitro Here, we explored whether this Sox11 manipulation similarly has stronger effects on RGC survival and optic nerve regeneration. In vitro, we found that non-SUMOylatable Sox11K91A leads to RGC death and suppresses axon outgrowth in primary neurons. We furthermore found that Sox11K91A more strongly promotes axon regeneration but also increases RGC death after optic nerve injury in vivo in the adult mouse. RNA sequence (RNA-seq) data showed that Sox11 and Sox11K91A increase the expression of key signaling pathway genes associated with axon growth and regeneration but downregulated Spp1 and Opn4 expression in RGC cultures, consistent with negatively regulating the survival of α-RGCs and ipRGCs. Thus, Sox11 and its SUMOylation site at K91 regulate gene expression, survival and axon growth in RGCs, and may be explored further as potential regenerative therapies for optic neuropathy.


Subject(s)
Neurodegenerative Diseases , Optic Nerve Injuries , Animals , Axons/metabolism , Cell Survival , Mice , Nerve Regeneration , Neurodegenerative Diseases/metabolism , Optic Nerve Injuries/metabolism , Protein Processing, Post-Translational , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism
5.
Toxicol Pathol ; 49(3): 544-554, 2021 04.
Article in English | MEDLINE | ID: mdl-32851936

ABSTRACT

A liquid chromatography-tandem mass spectrometry assay was developed and qualified for the multiplexed quantitation of a small molecule stimulator of soluble guanylate cyclase (sGC) and its target engagement biomarker, 3',5'-cyclic guanosine monophosphate (cGMP), in ocular tissues and plasma from a single surrogate matrix calibration curve. A surrogate matrix approach was used in this assay due to the limited quantities of blank ocular matrices in a discovery research setting. After optimization, the assay showed high accuracy, precision, and recovery as well as parallelism between the surrogate matrix and the biological matrices (rabbit plasma, vitreous, and retina-choroid). This assay provided pharmacokinetic and target engagement data after intravitreal administration of the sGC stimulator. The nitric oxide-sGC-cGMP pathway is a potential target to address glaucoma. Increasing sGC-mediated production of cGMP could improve aqueous humor outflow and ocular blood flow. The sGC stimulator showed dose-dependent exposure in rabbit vitreous, retina-choroid, and plasma. The cGMP exhibited a delayed yet sustained increase in vitreous humor but not retina-choroid. Multiplexed measurement of both pharmacokinetic and target engagement analytes reduced animal usage and provided improved context for interpreting PK and PD relationships.


Subject(s)
Cyclic GMP , Guanylate Cyclase , Animals , Guanylate Cyclase/metabolism , Nitric Oxide , Rabbits , Signal Transduction , Soluble Guanylyl Cyclase/metabolism
6.
Bio Protoc ; 10(6)2020 Mar 20.
Article in English | MEDLINE | ID: mdl-32368566

ABSTRACT

In diseases such as glaucoma, the failure of retinal ganglion cell (RGC) neurons to survive or regenerate their optic nerve axons underlies partial and, in some cases, complete vision loss. Optic nerve crush (ONC) serves as a useful model not only of traumatic optic neuropathy but also of glaucomatous injury, as it similarly induces RGC cell death and degeneration. Intravitreal injection of adeno-associated virus serotype 2 (AAV2) has been shown to specifically and efficiently transduce RGCs in vivo and has thus been proposed as an effective means of gene delivery for the treatment of glaucoma. Indeed, we and others routinely use AAV2 to study the mechanisms that promote neuroprotection and axon regeneration in RGCs following ONC. Herein, we describe a step-by-step protocol to assay RGC survival and regeneration in mice following AAV2-mediated transduction and ONC injury including 1) intravitreal injection of AAV2 viral vectors, 2) optic nerve crush, 3) cholera-toxin B (CTB) labeling of regenerating axons, 4) optic nerve clearing, 5) flat mount retina immunostaining, and 6) quantification of RGC survival and regeneration. In addition to providing all the materials and procedural details necessary to execute this protocol, we highlight its advantages over other similar published approaches and include useful tips to ensure its faithful reproduction in any modern laboratory.

7.
Sci Rep ; 10(1): 3375, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32099056

ABSTRACT

Optic neuropathy is a major cause of irreversible blindness worldwide, and no effective treatment is currently available. Secondary degeneration is believed to be the major contributor to retinal ganglion cell (RGC) death, the endpoint of optic neuropathy. Partial optic nerve transection (pONT) is an established model of optic neuropathy. Although the mechanisms of primary and secondary degeneration have been delineated in this model, until now how this is influenced by therapy is not well-understood. In this article, we describe a clinically translatable topical, neuroprotective treatment (recombinant human nerve growth factor, rh-NGF) predominantly targeting secondary degeneration in a pONT rat model. Topical application of rh-NGF twice daily for 3 weeks significantly improves RGC survival as shown by reduced RGC apoptosis in vivo and increased RGC population in the inferior retina, which is predominantly affected in this model by secondary degeneration. Topical rh-NGF also promotes greater axonal survival and inhibits astrocyte activity in the optic nerve. Collectively, these results suggest that topical rh-NGF exhibits neuroprotective effects on retinal neurons via influencing secondary degeneration process. As topical rh-NGF is already involved in early clinical trials, this highlights its potential in multiple indications in patients, including those affected by glaucomatous optic neuropathy.


Subject(s)
Apoptosis/drug effects , Nerve Growth Factor/pharmacology , Neuroprotective Agents/pharmacology , Administration, Topical , Animals , Axons/metabolism , Cell Survival/drug effects , Disease Models, Animal , Drug Administration Schedule , Humans , Male , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Nerve Growth Factor/therapeutic use , Neuroprotective Agents/therapeutic use , Optic Nerve Injuries/drug therapy , Optic Nerve Injuries/pathology , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Retina/metabolism , Retina/pathology , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism
8.
Restor Neurol Neurosci ; 38(2): 131-140, 2020.
Article in English | MEDLINE | ID: mdl-31815704

ABSTRACT

BACKGROUND: Glaucoma, the number one cause of irreversible blindness, is characterized by the loss of retinal ganglion cells (RGCs), which do not regenerate in humans or mammals after cell death. Cell transplantation provides an opportunity to restore vision in glaucoma, or other optic neuropathies. Since transplanting primary RGCs from deceased donor tissues may not be feasible, stem cell-derived RGCs could provide a plausible alternative source of donor cells for transplant. OBJECTIVE: We define a robust chemically defined protocol to differentiate human embryonic stem cells (hESCs) into RGC-like neurons. METHODS: Human embryonic stem cell lines (H7-A81 and H9) and induced pluripotent stem cell (iPSC) were used for RGC differentiation. RGC immaturity was measured by calcium imaging against muscimol. Cell markers were detected by immunofluorescence staining and qRT-PCR. RGC-like cells were intravitreally injected to rat eye, and co-stained with RBPMS and human nuclei markers. All experiments were conducted at least three times independently. Data were analyzed by ANOVA with Tukey's test with P value of <0.05 considered statistically significant. RESULTS: We detected retinal progenitor markers Rx and Pax6 after 15 days of differentiation, and the expression of markers for RGC-specific differentiation (Brn3a and Brn3b), maturation (synaptophysin) and neurite growth (ß-III-Tubulin) after an additional 15 days. We further examined the physiologic differentiation of these hESC-derived RGC-like progeny to those differentiated in vitro from primary rodent retinal progenitor cells (RPCs) with calcium imaging, and found that both populations demonstrate the immature RGC-like response to muscimol, a GABAA receptor agonist. By one week after transplant to the adult rat eye by intravitreal injection, the human RGC-like cells successfully migrated into the ganglion cell layer. CONCLUSIONS: Our protocol provides a novel, short, and cost-effective approach for RGC differentiation from hESCs, and may broaden the scope for cell replacement therapy in RGC-related optic neuropathies such as glaucoma.


Subject(s)
Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Retina/cytology , Retinal Ganglion Cells/physiology , Cell Differentiation/physiology , Cell Transplantation/methods , Humans , Induced Pluripotent Stem Cells/cytology , Neurogenesis/physiology
9.
Sci Rep ; 9(1): 10669, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31337818

ABSTRACT

The process of mitochondrial fission-fusion has been implicated in diverse neuronal roles including neuronal survival, axon degeneration, and axon regeneration. However, whether increased fission or fusion is beneficial for neuronal health and/or axonal growth is not entirely clear, and is likely situational and cell type-dependent. In searching for mitochondrial fission-fusion regulating proteins for improving axonal growth within the visual system, we uncover that mitochondrial fission process 1,18 kDa (MTP18/MTFP1), a pro-fission protein within the CNS, is critical to maintaining mitochondrial size and volume under normal and injury conditions, in retinal ganglion cells (RGCs). We demonstrate that MTP18's expression is regulated by transcription factors involved in axonal growth, Kruppel-like factor (KLF) transcription factors-7 and -9, and that knockdown of MTP18 promotes axon growth. This investigation exposes MTP18's previously unexplored role in regulating mitochondrial fission, implicates MTP18 as a downstream component of axon regenerative signaling, and ultimately lays the groundwork for investigations on the therapeutic efficacy of MTP18 expression suppression during CNS axon degenerative events.


Subject(s)
Axons/metabolism , Membrane Proteins/metabolism , Mitochondrial Dynamics/physiology , Mitochondrial Proteins/metabolism , Nerve Regeneration/physiology , Neurons/metabolism , Animals , Membrane Proteins/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Nerve Crush , Neuronal Outgrowth/physiology , Optic Nerve Injuries/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
10.
J Neurosci ; 39(28): 5466-5480, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31097623

ABSTRACT

cAMP signaling is known to be critical in neuronal survival and axon growth. Increasingly the subcellular compartmentation of cAMP signaling has been appreciated, but outside of dendritic synaptic regulation, few cAMP compartments have been defined in terms of molecular composition or function in neurons. Specificity in cAMP signaling is conferred in large part by A-kinase anchoring proteins (AKAPs) that localize protein kinase A and other signaling enzymes to discrete intracellular compartments. We now reveal that cAMP signaling within a perinuclear neuronal compartment organized by the large multivalent scaffold protein mAKAPα promotes neuronal survival and axon growth. mAKAPα signalosome function is explored using new molecular tools designed to specifically alter local cAMP levels as studied by live-cell FRET imaging. In addition, enhancement of mAKAPα-associated cAMP signaling by isoform-specific displacement of bound phosphodiesterase is demonstrated to increase retinal ganglion cell survival in vivo in mice of both sexes following optic nerve crush injury. These findings define a novel neuronal compartment that confers cAMP regulation of neuroprotection and axon growth and that may be therapeutically targeted in disease.SIGNIFICANCE STATEMENT cAMP is a second messenger responsible for the regulation of diverse cellular processes including neuronal neurite extension and survival following injury. Signal transduction by cAMP is highly compartmentalized in large part because of the formation of discrete, localized multimolecular signaling complexes by A-kinase anchoring proteins. Although the concept of cAMP compartmentation is well established, the function and identity of these compartments remain poorly understood in neurons. In this study, we provide evidence for a neuronal perinuclear cAMP compartment organized by the scaffold protein mAKAPα that is necessary and sufficient for the induction of neurite outgrowth in vitro and for the survival of retinal ganglion cells in vivo following optic nerve injury.


Subject(s)
Axon Guidance , Cyclic AMP/metabolism , Retinal Ganglion Cells/metabolism , Signal Transduction , A Kinase Anchor Proteins/metabolism , Animals , Axons/metabolism , Axons/physiology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Female , Fluorescence Resonance Energy Transfer , Male , Mice , Phosphoric Diester Hydrolases/metabolism , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/physiology
11.
Invest Ophthalmol Vis Sci ; 59(7): 2736-2747, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29860460

ABSTRACT

Purpose: Adult central nervous system (CNS) neurons are unable to regenerate their axons after injury. Krüppel-like transcription factor (KLF) family members regulate intrinsic axon growth ability in vitro and in vivo, but mechanisms downstream of these transcription factors are not known. Methods: Purified retinal ganglion cells (RGCs) were transduced to express exogenous KLF9, KLF16, KLF7, or KLF11; microarray analysis was used to identify downstream genes, which were screened for effects on axon growth. Dual-specificity phosphatase 14 (Dusp14) was further studied using genetic (siRNA, shRNA) and pharmacologic (PTP inhibitor IV) manipulation to assess effects on neurite length in vitro and survival and regeneration in vivo after optic nerve crush in rats and mice. Results: By screening genes regulated by KLFs in RGCs, we identified Dusp14 as a critical gene target limiting axon growth and regeneration downstream of KLF9's ability to suppress axon growth in RGCs. The KLF9-Dusp14 pathway inhibited activation of mitogen-activated protein kinases normally critical to neurotrophic signaling of RGC survival and axon elongation. Decreasing Dusp14 expression or disrupting its function in RGCs increased axon growth in vitro and promoted survival and optic nerve regeneration after optic nerve injury in vivo. Conclusions: These results link intrinsic and extrinsic regulators of axon growth and suggest modulation of the KLF9-Dusp14 pathway as a potential approach to improve regeneration in the adult CNS after injury.


Subject(s)
Axons/physiology , Dual-Specificity Phosphatases/genetics , Gene Expression Regulation/physiology , Kruppel-Like Transcription Factors/genetics , Nerve Regeneration/physiology , Optic Nerve Injuries/physiopathology , Animals , Blotting, Western , Dependovirus/genetics , Female , Fluorescent Antibody Technique, Indirect , Male , Nerve Crush , Plasmids , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Retinal Ganglion Cells/metabolism , Transfection
12.
J Neurosci ; 37(40): 9632-9644, 2017 10 04.
Article in English | MEDLINE | ID: mdl-28871032

ABSTRACT

Neurons in the adult mammalian CNS decrease in intrinsic axon growth capacity during development in concert with changes in Krüppel-like transcription factors (KLFs). KLFs regulate axon growth in CNS neurons including retinal ganglion cells (RGCs). Here, we found that knock-down of KLF9, an axon growth suppressor that is normally upregulated 250-fold in RGC development, promotes long-distance optic nerve regeneration in adult rats of both sexes. We identified a novel binding partner, MAPK10/JNK3 kinase, and found that JNK3 (c-Jun N-terminal kinase 3) is critical for KLF9's axon-growth-suppressive activity. Interfering with a JNK3-binding domain or mutating two newly discovered serine phosphorylation acceptor sites, Ser106 and Ser110, effectively abolished KLF9's neurite growth suppression in vitro and promoted axon regeneration in vivo These findings demonstrate a novel, physiologic role for the interaction of KLF9 and JNK3 in regenerative failure in the optic nerve and suggest new therapeutic strategies to promote axon regeneration in the adult CNS.SIGNIFICANCE STATEMENT Injured CNS nerves fail to regenerate spontaneously. Promoting intrinsic axon growth capacity has been a major challenge in the field. Here, we demonstrate that knocking down Krüppel-like transcription factor 9 (KLF9) via shRNA promotes long-distance axon regeneration after optic nerve injury and uncover a novel and important KLF9-JNK3 interaction that contributes to axon growth suppression in vitro and regenerative failure in vivo These studies suggest potential therapeutic approaches to promote axon regeneration in injury and other degenerative diseases in the adult CNS.


Subject(s)
Axons/physiology , Brain/physiology , Kruppel-Like Transcription Factors/metabolism , Mitogen-Activated Protein Kinase 10/metabolism , Nerve Regeneration/physiology , Age Factors , Animals , Base Sequence , Cells, Cultured , Central Nervous System/physiology , Female , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mitogen-Activated Protein Kinase 10/genetics , Optic Nerve Injuries/genetics , Optic Nerve Injuries/metabolism , Organ Culture Techniques , Protein Binding/physiology , Rats , Retinal Ganglion Cells/physiology
13.
Dev Neurobiol ; 77(4): 419-437, 2017 04.
Article in English | MEDLINE | ID: mdl-26999672

ABSTRACT

Serotonin (5HT) is present in a subpopulation of amacrine cells, which form synapses with retinal ganglion cells (RGCs), but little is known about the physiological role of retinal serotonergic circuitry. We found that the 5HT receptor 2C (5HTR2C) is upregulated in RGCs after birth. Amacrine cells generate 5HT and about half of RGCs respond to 5HTR2C agonism with calcium elevation. We found that there are on average 83 5HT+ amacrine cells randomly distributed across the adult mouse retina, all negative for choline acetyltransferase and 90% positive for tyrosine hydroxylase. We also investigated whether 5HTR2C and 5HTR5A affect RGC neurite growth. We found that both suppress neurite growth, and that RGCs from the 5HTR2C knockout (KO) mice grow longer neurites. Furthermore, 5HTR2C is subject to post-transcriptional editing, and we found that only the edited isoform's suppressive effect on neurite growth could be reversed by a 5HTR2C inverse agonist. Next, we investigated the physiological role of 5HTR2C in the retina, and found that 5HTR2C KO mice showed increased amplitude on pattern electroretinogram. Finally, RGC transcriptional profiling and pathways analysis suggested partial developmental compensation for 5HTR2C absence. Taken together, our findings demonstrate that 5HTR2C regulates neurite growth and RGC activity and is necessary for normal amplitude of RGC response to physiologic stimuli, and raise the hypothesis that these functions are modulated by a subset of 5HT+/ChAT-/TH+ amacrine cells as part of retinal serotonergic circuitry. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 419-437, 2017.


Subject(s)
Amacrine Cells/physiology , Neurites/physiology , Neurogenesis/physiology , Receptor, Serotonin, 5-HT2C/physiology , Retinal Ganglion Cells/physiology , Serotonin 5-HT2 Receptor Agonists/pharmacology , Vision, Ocular/physiology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Serotonin
14.
Cell Death Dis ; 7(12): e2514, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27929541

ABSTRACT

The amyloid beta (Aß) pathway is strongly implicated in neurodegenerative conditions such as Alzheimer's disease and more recently, glaucoma. Here, we identify the α2 adrenergic receptor agonists (α2ARA) used to lower intraocular pressure can prevent retinal ganglion cell (RGC) death via the non-amyloidogenic Aß-pathway. Neuroprotective effects were confirmed in vivo and in vitro in different glaucoma-related models using α2ARAs brimonidine (BMD), clonidine (Clo) and dexmedetomidine. α2ARA treatment significantly reduced RGC apoptosis in experimental-glaucoma models by 97.7% and 92.8% (BMD, P<0.01) and 98% and 92.3% (Clo, P<0.01)) at 3 and 8 weeks, respectively. A reduction was seen in an experimental Aß-induced neurotoxicity model (67% BMD and 88.6% Clo, both P<0.01, respectively), and in vitro, where α2ARAs significantly (P<0.05) prevented cell death, under both hypoxic (CoCl2) and stress (UV) conditions. In experimental-glaucoma, BMD induced ninefold and 25-fold and 36-fold and fourfold reductions in Aß and amyloid precursor protein (APP) levels at 3 and 8 weeks, respectively, in the RGC layer, with similar results with Clo, and in vitro with all three α2ARAs. BMD significantly increased soluble APPα (sAPPα) levels at 3 and 8 weeks (2.1 and 1.6-fold) in vivo and in vitro with the CoCl2 and UV-light insults. Furthermore, treatment of UV-insulted cells with an sAPPα antibody significantly reduced cell viability compared with BMD-treated control (52%), co-treatment (33%) and untreated control (27%). Finally, we show that α2ARAs modulate levels of laminin and MMP-9 in RGCs, potentially linked to changes in Aß through APP processing. Together, these results provide new evidence that α2ARAs are neuroprotective through their effects on the Aß pathway and sAPPα, which to our knowledge, is the first description. Studies have identified the need for α-secretase activators and sAPPα-mimetics in neurodegeneration; α2ARAs, already clinically available, present a promising therapy, with applications not only to reducing RGC death in glaucoma but also other neurodegenerative processes involving Aß.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Amyloid beta-Peptides/metabolism , Brimonidine Tartrate/pharmacology , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Death/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Male , Mice, Inbred C57BL , Models, Biological , Rats , Receptors, Adrenergic, beta/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Solubility
16.
J Biol Chem ; 291(35): 18084-95, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27402841

ABSTRACT

Regenerative medicine holds great promise for the treatment of degenerative retinal disorders. Krüppel-like factors (KLFs) are transcription factors that have recently emerged as key tools in regenerative medicine because some of them can function as epigenetic reprogrammers in stem cell biology. Here, we show that KLF16, one of the least understood members of this family, is a POU4F2 independent transcription factor in retinal ganglion cells (RGCs) as early as embryonic day 15. When overexpressed, KLF16 inhibits RGC neurite outgrowth and enhances RGC growth cone collapse in response to exogenous ephrinA5 ligands. Ephrin/EPH signaling regulates RGC connectivity. The EphA5 promoter contains multiple GC- and GT-rich KLF-binding sites, which, as shown by ChIP-assays, bind KLF16 in vivo In electrophoretic mobility shift assays, KLF16 binds specifically to a single KLF site near the EphA5 transcription start site that is required for KLF16 transactivation. Interestingly, methylation of only six of 98 CpG dinucleotides within the EphA5 promoter blocks its transactivation by KLF16 but enables transactivation by KLF2 and KLF15. These data demonstrate a role for KLF16 in regulation of RGC neurite outgrowth and as a methylation-sensitive transcriptional regulator of EphA5 expression. Together, these data identify differential low level methylation as a novel mechanism for regulating KLF16-mediated EphA5 expression across the retina. Because of the critical role of ephrin/EPH signaling in patterning RGC connectivity, understanding the role of KLFs in regulating neurite outgrowth and Eph receptor expression will be vital for successful restoration of functional vision through optic nerve regenerative therapies.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Neurites/metabolism , Receptor, EphA5/biosynthesis , Response Elements/physiology , Retinal Ganglion Cells/metabolism , Signal Transduction/physiology , Transcriptional Activation/physiology , Animals , DNA Methylation , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Receptor, EphA5/genetics , Retinal Ganglion Cells/cytology , Transcription, Genetic/physiology
17.
Exp Eye Res ; 140: 65-74, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26297614

ABSTRACT

Death of retinal neural cells, namely retinal ganglion cells (RGCs), is a characteristic of several retinal neurodegenerative diseases. Although the role of adenosine A3 receptor (A3R) in neuroprotection is controversial, A3R activation has been reported to afford protection against several brain insults, with few studies in the retina. In vitro models (retinal neural and organotypic cultures) and animal models [ischemia-reperfusion (I-R) and partial optic nerve transection (pONT)] were used to study the neuroprotective properties of A3R activation against retinal neurodegeneration. The A3R selective agonist (2-Cl-IB-MECA, 1 µM) prevented apoptosis (TUNEL(+)-cells) induced by kainate and cyclothiazide (KA + CTZ) in retinal neural cultures (86.5 ± 7.4 and 37.2 ± 6.1 TUNEL(+)-cells/field, in KA + CTZ and KA + CTZ + 2-Cl-IB-MECA, respectively). In retinal organotypic cultures, 2-Cl-IB-MECA attenuated NMDA-induced cell death, assessed by TUNEL (17.3 ± 2.3 and 8.3 ± 1.2 TUNEL(+)-cells/mm(2) in NMDA and NMDA+2-Cl-IB-MECA, respectively) and PI incorporation (ratio DIV4/DIV2 3.3 ± 0.3 and 1.3 ± 0.1 in NMDA and NMDA+2-Cl-IB-MECA, respectively) assays. Intravitreal 2-Cl-IB-MECA administration afforded protection against I-R injury decreasing the number of TUNEL(+) cells by 72%, and increased RGC survival by 57%. Also, intravitreal administration of 2-Cl-IB-MECA inhibited apoptosis (from 449.4 ± 37.8 to 207.6 ± 48.9 annexin-V(+)-cells) and RGC loss (from 1.2 ± 0.6 to 8.1 ± 1.7 cells/mm) induced by pONT. This study demonstrates that 2-Cl-IB-MECA is neuroprotective to the retina, both in vitro and in vivo. Activation of A3R may have great potential in the management of retinal neurodegenerative diseases characterized by RGC death, as glaucoma and diabetic retinopathy, and ischemic diseases.


Subject(s)
Neuroprotection/physiology , Receptor, Adenosine A3/metabolism , Retinal Degeneration/prevention & control , Retinal Neurons/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A3 Receptor Agonists/pharmacology , Adenosine A3 Receptor Antagonists/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Survival , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Fluorescent Antibody Technique, Indirect , In Situ Nick-End Labeling , Intravitreal Injections , Male , N-Methylaspartate/toxicity , Optic Nerve Injuries/metabolism , Organ Culture Techniques , Rats , Rats, Wistar , Retina/drug effects , Retina/pathology , Retinal Degeneration/metabolism
18.
FASEB J ; 28(3): 1317-30, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24327606

ABSTRACT

Dimethyl sulfoxide (DMSO) is an important aprotic solvent that can solubilize a wide variety of otherwise poorly soluble polar and nonpolar molecules. This, coupled with its apparent low toxicity at concentrations <10%, has led to its ubiquitous use and widespread application. Here, we demonstrate that DMSO induces retinal apoptosis in vivo at low concentrations (5 µl intravitreally dosed DMSO in rat from a stock concentration of 1, 2, 4, and 8% v/v). Toxicity was confirmed in vitro in a retinal neuronal cell line, at DMSO concentrations >1% (v/v), using annexin V, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and AlamarBlue cell viability assays. DMSO concentrations >10% (v/v) have recently been reported to cause cellular toxicity through plasma membrane pore formation. Here, we show the mechanism by which low concentrations (2-4% DMSO) induce caspase-3 independent neuronal death that involves apoptosis-inducing factor (AIF) translocation from mitochondria to the nucleus and poly-(ADP-ribose)-polymerase (PARP) activation. These results highlight safety concerns of using low concentrations of DMSO as a solvent for in vivo administration and in biological assays. We recommend that methods other than DMSO are employed for solubilizing drugs but, where no alternative exists, researchers compute absolute DMSO final concentrations and include an untreated control group in addition to DMSO vehicle control to check for solvent toxicity.


Subject(s)
Dimethyl Sulfoxide/toxicity , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line , Dose-Response Relationship, Drug , Male , Rats , Retinal Ganglion Cells/drug effects
19.
Curr Opin Pharmacol ; 13(1): 123-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22995681

ABSTRACT

Apoptosis, or programmed cell death, plays a vital role in normal development and ageing. However, dysregulation of this process is responsible for many disease states including; cancer, autoimmune and neurodegeneration. For this reason, in vivo visualisation of apoptosis may prove a useful tool for both laboratory research and clinical diagnostics. Glaucoma comprises a distinctive group of chronic optic neuropathies, characterised by the progressive loss of retinal ganglion cells (RGCs). Early diagnosis of glaucoma remains a clear and unmet need. Recently, there have been significant advances in the detection of apoptosis in vivo using fluorescent probes to visualise single RGCs undergoing apoptosis, specifically DARC (Detection of Apoptotic Retinal Cells) [1] and capQ technology [2(••)].


Subject(s)
Apoptosis , Glaucoma/pathology , Retinal Ganglion Cells/pathology , Animals , Diagnostic Imaging , Humans , Ophthalmoscopy/methods
20.
PLoS One ; 6(12): e29583, 2011.
Article in English | MEDLINE | ID: mdl-22216322

ABSTRACT

3,4-Methylenedioxymethamphetamine (MDMA; ecstasy) is known to produce euphoric states, but may also cause adverse consequences in humans, such as hyperthermia and neurocognitive deficits. Although MDMA consumption has been associated with visual problems, the effects of this recreational drug in retinal physiology have not been addressed hitherto. In this work, we evaluated the effect of a single MDMA administration in the rat electroretinogram (ERG). Wistar rats were administered MDMA (15 mg/kg) or saline and ERGs were recorded before (Baseline ERG), and 3 h, 24 h, and 7 days after treatment. A high temperature (HT) saline-treated control group was also included. Overall, significantly augmented and shorter latency ERG responses were found in MDMA and HT groups 3 h after treatment when compared to Baseline. Twenty-four hours after treatment some of the alterations found at 3 h, mainly characterized by shorter latency, tended to return to Baseline values. However, MDMA-treated animals still presented increased scotopic a-wave and b-wave amplitudes compared to Baseline ERGs, which were independent of temperature elevation though the latter might underlie the acute ERG alterations observed 3 h after MDMA administration. Seven days after MDMA administration recovery from these effects had occurred. The effects seem to stem from specific changes observed at the a-wave level, which indicates that MDMA affects subacutely (at 24 h) retinal physiology at the outer retinal (photoreceptor/bipolar) layers. In conclusion, we have found direct evidence that MDMA causes subacute enhancement of the outer retinal responses (most prominent in the a-wave), though ERG alterations resume within one week. These changes in photoreceptor/bipolar cell physiology may have implications for the understanding of the subacute visual manifestations induced by MDMA in humans.


Subject(s)
Kidney/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Animals , Electroretinography , Kidney/physiology , Male , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...