Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 953
Filter
Add more filters

Publication year range
1.
Am J Pathol ; 194(7): 1317-1328, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38548269

ABSTRACT

Two major constituents of exfoliation material, fibrillin-1 and lysyl oxidase-like 1 (encoded by FBN1 and LOXL1), are implicated in exfoliation glaucoma, yet their individual contributions to ocular phenotype are minor. To test the hypothesis that a combination of FBN1 mutation and LOXL1 deficiency exacerbates ocular phenotypes, the pan-lysyl oxidase inhibitor ß-aminopropionitrile (BAPN) was used to treat adult wild-type (WT) mice and mice heterozygous for a missense mutation in Fbn1 (Fbn1C1041G/+) for 8 weeks and their eyes were examined. Although intraocular pressure did not change and exfoliation material was not detected in the eyes, BAPN treatment worsened optic nerve and axon expansion in Fbn1C1041G/+ mice, an early sign of axonal damage in rodent models of glaucoma. Disruption of elastic fibers was detected only in Fbn1C1041G/+ mice, which increased with BAPN treatment, as shown by histologic and immunohistochemical staining of the optic nerve pia mater. Transmission electron microscopy showed that Fbn1C1041G/+ mice had fewer microfibrils, smaller elastin cores, and a lower density of elastic fibers compared with WT mice in control groups. BAPN treatment led to elastin core expansion in both WT and Fbn1C1041G/+ mice, but an increase in the density of elastic fiber was confined to Fbn1C1041G/+ mice. LOX inhibition had a stronger effect on optic nerve and elastic fiber parameters in the context of Fbn1 mutation, indicating the Marfan mouse model with LOX inhibition warrants further investigation for exfoliation glaucoma pathogenesis.


Subject(s)
Aminopropionitrile , Disease Models, Animal , Fibrillin-1 , Marfan Syndrome , Optic Nerve , Protein-Lysine 6-Oxidase , Animals , Protein-Lysine 6-Oxidase/metabolism , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Marfan Syndrome/pathology , Marfan Syndrome/complications , Mice , Fibrillin-1/genetics , Aminopropionitrile/pharmacology , Optic Nerve/pathology , Optic Nerve/ultrastructure , Optic Nerve/drug effects , Elastic Tissue/pathology , Elastic Tissue/metabolism , Elastic Tissue/ultrastructure , Intraocular Pressure , Fibrillins/metabolism , Mice, Inbred C57BL , Amino Acid Oxidoreductases/metabolism , Amino Acid Oxidoreductases/antagonists & inhibitors , Amino Acid Oxidoreductases/genetics , Glaucoma/pathology , Microfilament Proteins/metabolism , Adipokines
2.
Vestn Oftalmol ; 140(2): 91-96, 2024.
Article in Russian | MEDLINE | ID: mdl-38742504

ABSTRACT

Optic nerve atrophy is a pathomorphological consequence of diseases of the peripheral neuron of the visual pathway, manifested as atrophy of nerve fibers of varying severity. The toxic effect of methanol is mainly associated with formic acid and formaldehyde, which suppress the cytochrome system, inhibit oxidative phosphorylation, and thereby cause a deficiency of adenosine triphosphoric acid, to which brain and retinal tissues are especially susceptible. When formiate accumulates, tissue respiration is disrupted, leading to pronounced tissue hypoxia. As a result of such methanol metabolism, metabolic acidosis occurs. Tissue hypoxia develops in the first few hours as a result of the action of formic acid on the respiratory enzyme chain at the cytochrome oxidase level. Hypoxia and, as a consequence, a decrease in energy supply lead to a disruption of biological oxidation and the development of apoptosis in the optic nerve fibers. Understanding the process of optic nerve atrophy development at the pathogenetic level in methyl alcohol intoxication will help make a correct early diagnosis and prescribe timely treatment.


Subject(s)
Methanol , Optic Nerve , Humans , Methanol/poisoning , Optic Nerve/pathology , Optic Nerve/drug effects , Optic Atrophy/etiology , Optic Atrophy/diagnosis , Optic Atrophy/chemically induced
3.
Arch Toxicol ; 96(2): 431-451, 2022 02.
Article in English | MEDLINE | ID: mdl-34988610

ABSTRACT

Methanol-induced optic neuropathy (Me-ION) is a serious condition that may result in long-term or irreversible visual impairment or even blindness secondary to damage and loss of function of the optic nerve and retina. Me-ION shows a tendency to occur as mass poisonings around the world with a clear predilection for poor societies in developing countries. The main mechanism underlying the molecular basis of Me-ION is the inhibition of the mitochondrial oxidative phosphorylation process through the binding of the toxic metabolite of methanol-formic acid-with the key enzyme of this process-cytochrome c oxidase. However, other mechanisms, including damage to the eye tissues by oxidative stress causing the intensification of the oxidative peroxidation process with the formation of cytotoxic compounds, as well as an increase in the synthesis of pro-inflammatory cytokines and influence on the expression of key proteins responsible for maintaining cell homeostasis, also play an important role in the pathogenesis of Me-ION. Histopathological changes in the eye tissues are mainly manifested as the degeneration of axons and glial cells of the optic nerve, often with accompanying damage of the retina that may involve all its layers. Despite the development of therapeutic approaches, persistent visual sequelae are seen in 30-40% of survivors. Thus, Me-ION continues to be an important problem for healthcare systems worldwide.


Subject(s)
Methanol/poisoning , Optic Nerve Diseases/chemically induced , Optic Nerve/drug effects , Animals , Axons/drug effects , Axons/pathology , Formates/toxicity , Humans , Methanol/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Optic Nerve/pathology , Optic Nerve Diseases/physiopathology , Oxidative Stress/drug effects , Retina/drug effects , Retina/pathology
4.
Int J Mol Sci ; 23(4)2022 Feb 14.
Article in English | MEDLINE | ID: mdl-35216208

ABSTRACT

Clinically, acute ischemic symptoms in the eyes are one of the main causes of vision loss, with the associated inflammatory response and oxidative stress being the key factors that cause injury. Nonarteritic anterior ischemic optic neuropathy (NAION) is the most common type of ischemic optic neuropathy (ION); however, there are still no effective or safe treatment options to date. In this study, we investigated the neuroprotective effects of n-butylidenephthalide (BP) treatment in an experimental NAION rodent model (rAION). BP (10 mg/kg) or PBS (control group) were administered on seven consecutive days in the rAION model. Rats were evaluated for visual function by flash visual evoked potentials (FVEPs) at 4 weeks after NAION induction. The retina and optic nerve were removed for histological examination after the rats were euthanized. The molecular machinery of BP treatment in the rAION model was analyzed using Western blotting. We discovered that BP effectively improves retinal ganglion cell survival rates by preventing apoptotic processes after AION induction and reducing the inflammatory response through which blood-borne macrophages infiltrate the optic nerve. In addition, BP significantly preserved the integrity of the myelin sheath in the rAION model, demonstrating that BP can prevent the development of demyelination. Our immunoblotting results revealed the molecular mechanism through which BP mitigates the neuroinflammatory response through inhibition of the NF-κB signaling pathway. Taken together, these results demonstrate that BP can be used as an exceptional neuroprotective agent for ischemic injury.


Subject(s)
Ischemia/drug therapy , Neuroprotective Agents/pharmacology , Optic Neuropathy, Ischemic/drug therapy , Phthalic Anhydrides/pharmacology , Retinal Ganglion Cells/drug effects , Animals , Apoptosis/drug effects , Disease Models, Animal , Evoked Potentials, Visual/drug effects , Inflammation/drug therapy , Macrophages/drug effects , Male , Optic Nerve/drug effects , Rats , Rats, Wistar , Retina/drug effects
5.
Mol Pharmacol ; 100(5): 491-501, 2021 11.
Article in English | MEDLINE | ID: mdl-34470776

ABSTRACT

The neurotrophin growth factors bind and activate two types of cell surface receptors: the tropomyosin receptor kinase (Trk) family and p75. TrkA, TrkB, and TrkC are bound preferentially by nerve growth factor, brain-derived neurotrophic factor, and neurotrophin 3 (NT3), respectively, to activate neuroprotective signals. The p75 receptors are activated by all neurotrophins, and paradoxically in neurodegenerative disease p75 is upregulated and mediates neurotoxic signals. To test neuroprotection strategies, we engineered NT3 to broadly activate Trk receptors (mutant D) or to reduce p75 binding (mutant RK). We also combined these features in a molecule that activates TrkA, TrkB, and TrkC but has reduced p75 binding (mutant DRK). In neurodegenerative disease mouse models in vivo, the DRK protein is a superior therapeutic agent compared with mutant D, mutant RK, and wild-type neurotrophins and protects a broader range of stressed neurons. This work rationalizes a therapeutic strategy based on the biology of each type of receptor, avoiding activation of p75 toxicity while broadly activating neuroprotection in stressed neuronal populations expressing different Trk receptors. SIGNIFICANCE STATEMENT: The neurotrophins nerve growth factor, brain-derived neurotrophic factor, and neurotrophin 3 each can activate a tropomyosin receptor kinase (Trk) A, TrkB, or TrkC receptor, respectively, and all can activate a p75 receptor. Trks and p75 mediate opposite signals. We report the engineering of a protein that activates all Trks, combined with low p75 binding, as an effective therapeutic agent in vivo.


Subject(s)
Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neuroprotection/physiology , Protein Engineering/methods , Receptor, trkA/metabolism , Receptors, Growth Factor/metabolism , Animals , Axotomy/adverse effects , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/genetics , Diabetic Neuropathies/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Neuroprotection/drug effects , Optic Nerve/drug effects , Optic Nerve/metabolism , Receptor, trkA/genetics , Receptors, Growth Factor/genetics
6.
Neurobiol Dis ; 150: 105259, 2021 03.
Article in English | MEDLINE | ID: mdl-33434618

ABSTRACT

Neuronal regeneration in the injured central nervous system is hampered by multiple extracellular proteins. These proteins exert their inhibitory action through interactions with receptors that are located in cholesterol rich compartments of the membrane termed lipid rafts. Here we show that cholesterol-synthesis inhibition prevents the association of the Neogenin receptor with lipid rafts. Furthermore, we show that cholesterol-synthesis inhibition enhances axonal growth both on inhibitory -myelin and -RGMa substrates. Following optic nerve injury, lowering cholesterol synthesis with both drugs and siRNA-strategies allows for robust axonal regeneration and promotes neuronal survival. Cholesterol inhibition also enhanced photoreceptor survival in a model of Retinitis Pigmentosa. Our data reveal that Lovastatin leads to several opposing effects on regenerating axons: cholesterol synthesis inhibition promotes regeneration whereas altered prenylation impairs regeneration. We also show that the lactone prodrug form of lovastatin has differing effects on regeneration when compared to the ring-open hydroxy-acid form. Thus the association of cell surface receptors with lipid rafts contributes to axonal regeneration inhibition, and blocking cholesterol synthesis provides a potential therapeutic approach to promote neuronal regeneration and survival in the diseased Central Nervous System. SIGNIFICANCE STATEMENT: Statins have been intensively used to treat high levels of cholesterol in humans. However, the effect of cholesterol inhibition in both the healthy and the diseased brain remains controversial. In particular, it is unclear whether cholesterol inhibition with statins can promote regeneration and survival following injuries. Here we show that late stage cholesterol inhibition promotes robust axonal regeneration following optic nerve injury. We identified distinct mechanisms of action for activated vs non-activated Lovastatin that may account for discrepancies found in the literature. We show that late stage cholesterol synthesis inhibition alters Neogenin association with lipid rafts, thereby i) neutralizing the inhibitory function of its ligand and ii) offering a novel opportunity to promote CNS regeneration and survival following injuries.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Nerve Regeneration/drug effects , Neurons/drug effects , Optic Nerve/drug effects , Animals , Anticholesteremic Agents/pharmacology , Axons/drug effects , Axons/pathology , Cell Survival , Chick Embryo , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Myelin Sheath , Neurons/metabolism , Optic Nerve/metabolism , Optic Nerve/pathology , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Photoreceptor Cells , Prenylation , Prodrugs , Rats , Retina , Retinitis Pigmentosa , trans-1,4-Bis(2-chlorobenzaminomethyl)cyclohexane Dihydrochloride/pharmacology
7.
Neurobiol Dis ; 159: 105492, 2021 11.
Article in English | MEDLINE | ID: mdl-34478849

ABSTRACT

Repulsive guidance molecule A (RGMa) is a potent inhibitor of axonal growth and a regulator of neuronal cell death. It is up-regulated following neuronal injury and accumulates in chronic neurodegenerative diseases. Neutralizing RGMa has the potential to promote neuroregeneration and neuroprotection. Previously we reported that a rat anti-N terminal RGMa (N-RGMa) antibody r5F9 and its humanized version h5F9 (ABT-207) promote neuroprotection and neuroregeneration in preclinical neurodegenerative disease models. However, due to its cross-reactivity to RGMc/hemojuvelin, ABT-207 causes iron accumulation in vivo, which could present a safety liability. Here we report the generation and characterization of a novel RGMa-selective anti-N-RGMa antibody elezanumab, which is currently under Phase 2 clinical evaluation in multiple disease indications. Elezanumab, a human monoclonal antibody generated by in vitro PROfusion mRNA display technology, competes with ABT-207 in binding to N-RGMa but lacks RGMc cross-reactivity with no impact on iron metabolism. It neutralizes repulsive activity of soluble RGMa in vitro and blocks membrane RGMa mediated BMP signaling. In the optic nerve crush and optic neuritis models, elezanumab promotes axonal regeneration and prevents retinal nerve fiber layer degeneration. In the spinal targeted experimental autoimmune encephalomyelitis (EAE) model, elezanumab promotes axonal regeneration and remyelination, decreases inflammatory lesion area and improves functional recovery. Finally, in the mouse cuprizone model, elezanumab reduces demyelination, which is consistent with its inhibitory effect on BMP signaling. Taken together, these preclinical data demonstrate that elezanumab has neuroregenerative and neuroprotective activities without impact on iron metabolism, thus providing a compelling rationale for its clinical development in neurodegenerative diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , GPI-Linked Proteins , Nerve Regeneration , Nerve Tissue Proteins , Neuroprotection , Optic Nerve Injuries , Optic Nerve , Optic Neuritis , Recovery of Function , Retina , Animals , Mice , Cuprizone/toxicity , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/physiopathology , GPI-Linked Proteins/antagonists & inhibitors , Monoamine Oxidase Inhibitors/toxicity , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Nerve Tissue Proteins/antagonists & inhibitors , Neuroprotection/drug effects , Optic Nerve/drug effects , Optic Nerve/physiology , Optic Nerve Injuries/physiopathology , Optic Neuritis/physiopathology , Recovery of Function/drug effects , Recovery of Function/physiology , Retina/drug effects , Surface Plasmon Resonance
8.
Neurobiol Dis ; 147: 105146, 2021 01.
Article in English | MEDLINE | ID: mdl-33122075

ABSTRACT

Pathological accumulations of amyloid-beta (Aß) peptide are found in retina early in Alzheimer's disease, yet its effects on retinal neuronal structure remain unknown. To investigate this, we injected fibrillized Aß1-42 protein into the eye of adult C57BL/6 J mice and analyzed the retina, optic nerve (ON), and the superior colliculus (SC), the primary retinal target in mice. We found that retinal Aß exposure stimulated microglial activation and retinal ganglion cell (RGC) loss as early as 1-week post-injection. Pathology was not limited to the retina, but propagated into other areas of the central nervous system. Microgliosis spread throughout the retinal projection (retina, ON, and SC), with multiplex protein quantitation demonstrating an increase in endogenously produced Aß in the ON and SC corresponding to the injected retinas. Surprisingly, this pathology spread to the opposite side, with unilateral Aß eye injections driving increased Aß levels, neuroinflammation, and RGC death in the opposite, un-injected retinal projection. As Aß-mediated microglial activation has been shown to propagate Aß pathology, we also investigated the role of the Aß-binding microglial scavenger receptor CD36 in this pathology. Transgenic mice lacking the CD36 receptor were resistant to Aß-induced inflammation and RGC death up to 2 weeks following exposure. These results indicate that Aß pathology drives regional neuropathology in the retina and does not remain isolated to the affected eye, but spreads throughout the nervous system. Further, CD36 may serve as a promising target to prevent Aß-mediated inflammatory damage.


Subject(s)
Amyloid beta-Protein Precursor/toxicity , Gliosis/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Animals , CD36 Antigens/metabolism , Female , Humans , Intravitreal Injections , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Optic Nerve/drug effects , Optic Nerve/pathology , Retina/drug effects , Retina/pathology , Superior Colliculi/drug effects , Superior Colliculi/pathology
9.
BMC Anesthesiol ; 21(1): 30, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33504329

ABSTRACT

BACKGROUND: The results of studies on changes in intracranial pressure in patients undergoing laparoscopic surgery are inconsistent. Meanwhile, previous neurosurgery studies have suggested that propofol and sevoflurane have inconsistent effects on cerebral blood flow and cerebrovascular self-regulation. The purpose of this study is to compare changes in the optic nerve sheath diameter in patients undergoing laparoscopic gynecological surgery under anesthetic maintenance with propofol versus sevoflurane. METHODS: This study included 110 patients undergoing laparoscopic gynecological surgery with an estimated operative time of more than 2 h under general anesthesia. The study was a randomized controlled study. The optic nerve sheath diameter (ONSD) at various time points was measured by ultrasound, including when the patients entered the operating room (Tawake), after successful anesthesia induction and endotracheal intubation (Tinduction), when the body position was adjusted to the Trendelenburg position and the CO2 pneumoperitoneum pressure reached 14 mmHg, which was recorded as T0. Then, measurements were conducted every 15 min for the first 1 h and then once every hour until the end of the surgery (T15, T30, T45, T1h, T2h …), after the end of surgery and the tracheal tube was removed (Tend), and before the patients were transferred to the ward (Tpacu). RESULTS: A significant difference in optic nerve sheath diameter was found between two groups at T15, T30, T45 (4.64 ± 0.48 mm and 4.50 ± 0.29 mm, respectively, p = 0.031;4.77 ± 0.45 mm and 4.62 ± 0.28 mm, respectively, p = 0.036;4.84 ± 0.46 mm and 4.65 ± 0.30 mm, respectively, p = 0.012), while there was no significant difference at Tawake and other time points. CONCLUSION: During laparoscopic gynecological surgery lasting more than 2 h, the optic nerve sheath diameter was slightly larger in the propofol group than that in the sevoflurane group in the first 45 min. No significant difference was observed between the two groups 1 h after surgery. TRIAL REGISTRATION: clinicaltrials.gov, NCT03498235 . Retrospectively registered 1 March 2018. The manuscript adheres to CONSORT guidelines.


Subject(s)
Gynecologic Surgical Procedures/methods , Optic Nerve/anatomy & histology , Optic Nerve/drug effects , Propofol/pharmacology , Sevoflurane/pharmacology , Adult , Anesthetics, Inhalation/pharmacology , Anesthetics, Intravenous/pharmacology , Female , Humans , Laparoscopy
10.
Int J Mol Sci ; 22(5)2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33671012

ABSTRACT

The need for remyelinating drugs is essential for healing disabling diseases such as multiple sclerosis (MS). One of the reasons for the lack of this class of therapies is the impossibility to monitor remyelination in vivo, which is of utmost importance to perform effective clinical trials. Here, we show how optical coherence tomography (OCT), a cheap and non-invasive technique commonly used in ophthalmology, may be used to assess remyelination in vivo in MS patients. Our pioneer approach validates OCT as a technique to study remyelination of the optic nerve and reflects what is occurring in non-accessible central nervous system (CNS) structures, like the spinal cord. In this study we used the orally bioavailable small molecule VP3.15, confirming its therapeutical potential as a neuroprotective, anti-inflammatory, and probably remyelinating drug for MS. Altogether, our results confirm the usefulness of OCT to monitor the efficacy of remyelinating therapies in vivo and underscore the relevance of VP3.15 as a potential disease modifying drug for MS therapy.


Subject(s)
Multiple Sclerosis/drug therapy , Optic Nerve/drug effects , Remyelination , Small Molecule Libraries/pharmacology , Tomography, Optical Coherence/methods , Animals , Female , Mice , Mice, Inbred C57BL , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/pathology , Neuroprotection , Optic Nerve/diagnostic imaging , Optic Nerve/pathology
11.
Int J Mol Sci ; 22(14)2021 Jul 17.
Article in English | MEDLINE | ID: mdl-34299278

ABSTRACT

During the pathogenesis of glaucoma, optic nerve (ON) axons become continuously damaged at the optic nerve head (ONH). This often is associated with reactive astrocytes and increased transforming growth factor (TGF-ß) 2 levels. In this study we tested the hypothesis if the presence or absence of decorin (DCN), a small leucine-rich proteoglycan and a natural inhibitor of several members of the TGF family, would affect the expression of the TGF-ßs and connective tissue growth factor (CTGF/CCN2) in human ONH astrocytes and murine ON astrocytes. We found that DCN is present in the mouse ON and is expressed by human ONH and murine ON astrocytes. DCN expression and synthesis was significantly reduced after 24 h treatment with 3 nM CTGF/CCN2, while treatment with 4 pM TGF-ß2 only reduced expression of DCN significantly. Conversely, DCN treatment significantly reduced the expression of TGF-ß1, TGF-ß2 and CTGF/CCN2 vis-a-vis untreated controls. Furthermore, DCN treatment significantly reduced expression of fibronectin (FN) and collagen IV (COL IV). Notably, combined treatment with DCN and triciribine, a small molecule inhibitor of protein kinase B (AKT), attenuated effects of DCN on CTGF/CCN2, TGF-ß1, and TGF-ß2 mRNA expression. We conclude (1) that DCN is an important regulator of TGF-ß and CTGF/CCN2 expression in astrocytes of the ON and ONH, (2) that DCN thereby regulates the expression of extracellular matrix (ECM) components and (3) that DCN executes its negative regulatory effects on TGF-ß and CTGF/CCN2 via the pAKT/AKT signaling pathway in ON astrocytes.


Subject(s)
Astrocytes/metabolism , Decorin/pharmacology , Extracellular Matrix Proteins/metabolism , Glaucoma/pathology , Oncogene Protein v-akt/metabolism , Optic Nerve/metabolism , Transforming Growth Factor beta2/antagonists & inhibitors , Animals , Astrocytes/drug effects , Cells, Cultured , Disease Models, Animal , Female , Glaucoma/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Optic Nerve/drug effects , Signal Transduction
12.
Int J Mol Sci ; 22(22)2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34830387

ABSTRACT

Glaucoma is an optic neuropathy in which the degeneration of retinal ganglion cells (RGCs) results in irreversible vison loss. Therefore, neuroprotection of RGCs from glaucomatous afflictions is crucial for glaucoma treatment. In this study, we aimed to investigate the beneficial effects of statins in the protection of RGCs using a rat model. Glaucomatous injury was induced in rats by chronic ocular hypertension (OHT) achieved after performing a circumlimbal suture. The rats were given either statins such as simvastatin and atorvastatin or a solvent weekly for 6 weeks. Retina sections underwent hematoxylin and eosin, Brn3a, or cleaved casepase-3 staining to evaluate RGC survival. In addition, modulation of glial activation was assessed. While the retinas without statin treatment exhibited increased RGC death due to chronic OHT, statins promoted the survival of RGCs and reduced apoptosis. Statins also suppressed chronic OHT-mediated glial activation in the retina. Our results demonstrate that statins exert neuroprotective effects in rat retinas exposed to chronic OHT, which may support the prospect of statins being a glaucoma treatment.


Subject(s)
Glaucoma/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Ocular Hypertension/drug therapy , Retinal Degeneration/drug therapy , Animals , Disease Models, Animal , Glaucoma/genetics , Glaucoma/pathology , Humans , Intraocular Pressure/drug effects , Neuroprotection/genetics , Neuroprotective Agents/pharmacology , Ocular Hypertension/genetics , Ocular Hypertension/pathology , Optic Nerve/drug effects , Optic Nerve/pathology , Optic Nerve Diseases/drug therapy , Optic Nerve Diseases/genetics , Optic Nerve Diseases/pathology , Rats , Retina/drug effects , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Transcription Factor Brn-3A/chemistry , Transcription Factor Brn-3A/isolation & purification
13.
Cutan Ocul Toxicol ; 40(1): 1-6, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33121287

ABSTRACT

AIM: To investigate the effect of taxifolin on cisplatin-induced oxidative and proinflammatory optic nerve damage in rats. METHODS: A total of 18 albino Wistar male rats were assigned into 3 groups, as follows; Group 1: Control group, Group 2: Only cisplatin administered group for 14 days (Cisplatin group), and Group 3: Taxifolin + cisplatin administered group for 14 days (CIS + TAX group). Serum malondialdehyde (MDA), total Glutathione (tGSH), Nuclear Factor-Kappa B (NF-ƘB), Total Oxidative Status (TOS) and Total Antioxidant Status (TAS) levels were collected from the left eyes of rats. Rats' right eyes were enucleated for histopathological evaluations of optic nerves. RESULTS: NF-ƘB, MDA and TOS levels were statistically significantly higher (p < 0.001) in cisplatin group when compared to other 2 groups, the tGSH and TAS levels of which were statistically significantly lower (p < 0.001). Regarding these parameters, in cisplatin group NF-ƘB, MDA and TOS levels were statistically significantly increased with cisplatin administration and giving taxifolin concomitantly with cisplatin prevented this elevation. On the other hand, tGSH and TAS levels were statistically significantly decreased with cisplatin administration and routine simultaneous application of taxifolin with cisplatin prevented this decrease. In histopathological findings, haemorrhage was observed in the perineum of the injured optic nerves in the cisplatin treated group. And also edoema and degeneration in nerve fascicles in damaged optic nerves were seen in the cisplatin group. In the taxifolin treated group histopathological examinations were close to normal appearance, except mild edoema in nerve fascicles. CONCLUSION: Cisplatin causes oxidative stress on the rat optic nerves, and these changes lead to significant histopathological damage. Taxifolin, which we used to prevent oxidative damage to the optic nerves caused by cisplatin, has been emphasized as a powerful antioxidant agent in many previous scientific investigations. Concomitant administration of taxifolin may prevent these adverse effects of cisplatin, as well as histopathological damage. Further studies are needed to fully determine the effects of cisplatin and taxifolin on the eye.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Cisplatin/adverse effects , Optic Nerve Diseases/drug therapy , Optic Nerve/drug effects , Quercetin/analogs & derivatives , Animals , Disease Models, Animal , Male , Optic Nerve/pathology , Optic Nerve Diseases/chemically induced , Oxidative Stress/drug effects , Oxidative Stress/immunology , Quercetin/administration & dosage , Rats
14.
Cutan Ocul Toxicol ; 40(3): 263-267, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34114905

ABSTRACT

AIM: To investigate the effects of resveratrol on oxidative stress and inflammation parameters and histological alterations in cisplatin-induced optic nerve damage in a mouse model. MATERIAL AND METHOD: Thirty-six albino Wistar male rats were divided into three groups as control, 5 mg/kg cisplatin-administered (Cis) and 5 mg/kg cisplatin + 25 mg/kg resveratrol-administered (Cis + Res) animals. At the end of the experimental period, the rats were sacrificed with high-dose (50 mg/kg) thiopental sodium, and their optic nerves were dissected. Malondialdehyde (MDA), total glutathione (tGSH), total oxidant status (TOS), total antioxidant status (TAS), tumour necrosis factor alpha (TNF-α), nuclear factor kappa B (NF-KB) levels, and histopathological findings were assessed using the optic nerve tissues. RESULTS: In the Cis + Res group, the MDA, TOS, OSI, TNF-a and NFK-B levels were significantly lower and the tGSH and TAS levels were significantly higher compared with the Cis group (P = 0.001). In histological evaluations, there were dilated and congested blood vessels, destruction, oedema, degeneration, haemorrhage, and proliferating capillaries indicating the presence of inflammation and damage only in the Cis-administered group. However, in the Cis + Res group, the histological findings were very similar to the healthy controls. CONCLUSION: Resveratrol is a promising neuroprotective agent for cisplatin-induced optic nerve toxicity with its anti-oxidant and anti-inflammatory effects. Further investigations are needed to evaluate the possible therapeutic effects on other optic nerve toxicities.


Subject(s)
Cisplatin/adverse effects , Neuroprotective Agents/administration & dosage , Optic Nerve Injuries/drug therapy , Optic Nerve/drug effects , Resveratrol/administration & dosage , Animals , Disease Models, Animal , Humans , Male , Optic Nerve/immunology , Optic Nerve/pathology , Optic Nerve Injuries/chemically induced , Optic Nerve Injuries/immunology , Optic Nerve Injuries/pathology , Oxidative Stress/drug effects , Oxidative Stress/immunology , Rats
15.
J Neurochem ; 153(6): 693-709, 2020 06.
Article in English | MEDLINE | ID: mdl-32031240

ABSTRACT

Autoimmune optic neuritis (AON), a model of multiple sclerosis-associated optic neuritis, is accompanied by degeneration of retinal ganglion cells (RGCs) and optic nerve demyelination and axonal loss. In order to investigate the role of N-methyl-d-aspartate (NMDA) receptors in mediating RGC degeneration, upstream changes in the optic nerve actin cytoskeleton and associated deterioration in visual function, we induced AON in Brown Norway rats by immunization with myelin oligodendrocyte glycoprotein. Subsequently, visual acuity was assessed by recording visual evoked potentials and electroretinograms prior to extraction of optic nerves for western blot analysis and retinas for quantification of RGCs. As previously reported, in Brown Norway rats RGC degeneration is observed prior to onset of immune cell infiltration and demyelination of the optic nerves. However, within the optic nerve, destabilization of the actin cytoskeleton could be seen as indicated by an increase in the globular to filamentous actin ratio. Interestingly, these changes could be mimicked by intravitreal injection of glutamate, and similarly blocked by application of the NMDA receptor blocker MK-801, leading us to propose that prior to optic nerve lesion formation, NMDA receptor activation within the retina leads to retinal calcium accumulation, actin destabilization within the optic nerve as well as a deterioration of visual acuity during AON.


Subject(s)
Optic Neuritis/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Retina/metabolism , Animals , Dizocilpine Maleate/pharmacology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Evoked Potentials, Visual/drug effects , Evoked Potentials, Visual/physiology , Excitatory Amino Acid Antagonists/pharmacology , Female , Myelin-Oligodendrocyte Glycoprotein/toxicity , Optic Nerve/drug effects , Optic Nerve/immunology , Optic Nerve/metabolism , Optic Neuritis/chemically induced , Optic Neuritis/immunology , Rats , Rats, Inbred BN , Receptors, N-Methyl-D-Aspartate/immunology , Retina/drug effects , Retina/immunology
16.
Neurobiol Dis ; 134: 104695, 2020 02.
Article in English | MEDLINE | ID: mdl-31778813

ABSTRACT

Our goal was to investigate the neuroprotective effects of galantamine in a mouse model of blast-induced indirect traumatic optic neuropathy (bITON). Galantamine is an FDA-approved acetylcholinesterase inhibitor used to treat mild-moderate Alzheimer's disease. We exposed one eye of an anesthetized mouse to repeat bursts of over-pressurized air to induce traumatic optic neuropathy. Mice were given regular or galantamine-containing water (120 mg/L) ad libitum, beginning immediately after blast and continuing for one month. Electroretinograms and visual evoked potentials were performed just prior to endpoint collection. Histological and biochemical assessments were performed to assess activation of sterile inflammation, axon degeneration, and synaptic changes. Galantamine treatment mitigated visual function deficits induced by our bITON model via preservation of the b-wave of the electroretinogram and the N1 of the visual evoked potential. We also observed a reduction in axon degeneration in the optic nerve as well as decreased rod bipolar cell dendritic retraction. Galantamine also showed anti-inflammatory and antioxidant effects. Galantamine may be a promising treatment for blast-induced indirect traumatic optic neuropathy as well as other optic neuropathies.


Subject(s)
Axons/drug effects , Cholinesterase Inhibitors/administration & dosage , Evoked Potentials, Visual/drug effects , Galantamine/administration & dosage , Neuroprotective Agents/administration & dosage , Optic Nerve Injuries/pathology , Optic Nerve Injuries/physiopathology , Synapses/drug effects , Acetylcholinesterase/analysis , Administration, Oral , Animals , Axons/pathology , Male , Mice, Inbred C57BL , Optic Nerve/drug effects , Optic Nerve/pathology , Optic Nerve Injuries/complications , Retina/drug effects , Retina/pathology , Synapses/pathology
17.
Mol Vis ; 26: 135-149, 2020.
Article in English | MEDLINE | ID: mdl-32180679

ABSTRACT

Purpose: To analyze whether activation of endogenous wingless (Wnt)/ß-catenin signaling in Müller cells is involved in protection of retinal ganglion cells (RGCs) following excitotoxic damage. Methods: Transgenic mice with a tamoxifen-dependent ß-catenin deficiency in Müller cells were injected with N-methyl-D-aspartate (NMDA) into the vitreous cavity of one eye to induce excitotoxic damage of the RGCs, while the contralateral eye received PBS only. Retinal damage was quantified by counting the total number of RGC axons in cross sections of optic nerves and measuring the thickness of the retinal layers on meridional sections. Then, terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay was performed to identify apoptotic cells in retinas of both genotypes. Western blot analyses to assess the level of retinal ß-catenin and real-time RT-PCR to quantify the retinal expression of neuroprotective factors were performed. Results: Following NMDA injection of wild-type mice, a statistically significant increase in retinal ß-catenin protein levels was observed compared to PBS-injected controls, an effect that was blocked in mice with a Müller cell-specific ß-catenin deficiency. Furthermore, in mice with a ß-catenin deficiency in Müller cells, NMDA injection led to a statistically significant decrease in RGC axons as well as a substantial increase in TUNEL-positive cells in the RGC layer compared to the NMDA-treated controls. Moreover, in the retinas of the control mice a NMDA-mediated statistically significant induction of leukemia inhibitory factor (Lif) mRNA was detected, an effect that was substantially reduced in mice with a ß-catenin deficiency in Müller cells. Conclusions: Endogenous Wnt/ß-catenin signaling in Müller cells protects RGCs against excitotoxic damage, an effect that is most likely mediated via the induction of neuroprotective factors, such as Lif.


Subject(s)
Ependymoglial Cells/metabolism , Optic Nerve/metabolism , Retina/metabolism , Retinal Ganglion Cells/metabolism , Tamoxifen/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Apoptosis/drug effects , Axons/drug effects , Axons/metabolism , Ependymoglial Cells/drug effects , In Situ Nick-End Labeling , Leukemia Inhibitory Factor/metabolism , Mice , Mice, Transgenic , N-Methylaspartate/toxicity , Optic Nerve/drug effects , Retina/drug effects , Retina/pathology , Retinal Ganglion Cells/drug effects , Wnt Signaling Pathway/genetics , beta Catenin/deficiency
18.
J Recept Signal Transduct Res ; 40(2): 97-108, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32019426

ABSTRACT

Background: Cisplatin (CiSP), a chemotherapeutic agent, is widely used to treat several types of cancers. However, its clinical use is limited due to adverse side effects caused by excessive production of reactive oxygen species (ROS) and death of neurons. The transient receptor potential (TRP) melastatin 2 (TRPM2) cation channel is activated by ADP-ribose (ADPR) and ROS. The protective effect of curcumin (CURCU) against CiSP-induced apoptosis and mitochondrial ROS through inhibition of TRP channels in several types of neuron except optic nerve, was recently reported. The aim of the current study is to clarify the protective effect of CURCU on CiSP-induced mitochondrial oxidative injury and TRPM2 activation in the mice optic nerve and SH-SY5Y human derived neuronal cells.Material and methods: The SH-SY5Y cells and mice were divided into four groups: Control, CURCU, CiSP, and CURCU + CiSP. The mice were treated for 14 days and the cells were incubated with CiSP and CURCU for 24 h.Results: CURCU and PARP-1 inhibitor (PJ34) treatments ameliorated CiSP-induced mitochondrial membrane depolarization, mitochondrial and cytosolic ROS levels and neuronal death in the optic nerve. In the patch-clamp of SH-SY5Y cells and laser confocal microscopy experiments of optic nerve, CURCU and TRPM2 blocker treatments also decreased ADPR-induced TRPM2 currents and cytosolic free calcium ion (Ca2+) concentration, suggesting a suppression of Ca2+ influx and neuronal death.Conclusion: CURCU prevents CiSP-induced optic nerve oxidative injury and cell death by suppressing mitochondrial ROS production via regulating TRPM2 signaling pathways. CURCU may serve as a potential therapeutic target against CiSP-induced toxicity in the optic nerve of CiSP-treated patients.


Subject(s)
Curcumin/pharmacology , Optic Nerve Injuries/drug therapy , Optic Nerve/drug effects , Oxidative Stress/drug effects , TRPM Cation Channels/genetics , Adenosine Diphosphate Ribose/genetics , Animals , Apoptosis/drug effects , Cisplatin/toxicity , Humans , Mice , Mitochondria/drug effects , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/genetics , Optic Nerve/pathology , Optic Nerve Injuries/chemically induced , Optic Nerve Injuries/genetics , Optic Nerve Injuries/pathology , Patch-Clamp Techniques , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
19.
Exp Eye Res ; 193: 107961, 2020 04.
Article in English | MEDLINE | ID: mdl-32045598

ABSTRACT

Glaucoma is a progressive neurodegenerative process affecting the retinal ganglion cells (RGCs) and the optic nerve. Oxidative stress has been implicated in glaucoma pathogenesis, and iron is a potent generator of oxidative stress. The oral iron chelator deferiprone (DFP) is protective against retinal degenerations associated with oxidative stress. To test whether DFP could be protective in glaucoma, we used microbead injections to induce elevated intraocular pressure (IOP) in a cohort of 3-month old C57BL/6J mice. One eye of each animal was injected with magnetic microbeads resulting in ocular hypertension for >7 weeks while the fellow eye was injected with saline and served as a normotensive internal control. While half of the cohort received oral DFP (1 mg/ml in the drinking water), the other half did not and served as controls. After 8 weeks, Brn3a immunolabeling of flat-mounted retinas was used for manual RGC quantification. Axon counts were obtained from thin sections of optic nerves using the AxonJ plugin for ImageJ. DFP administration was protective against RGC and optic nerve loss in the setting of elevated IOP. These results suggest that iron chelation by DFP may provide glaucoma neuroprotection.


Subject(s)
Deferiprone/administration & dosage , Glaucoma/complications , Optic Nerve/pathology , Retinal Degeneration/prevention & control , Retinal Ganglion Cells/pathology , Administration, Oral , Animals , Disease Models, Animal , Female , Glaucoma/drug therapy , Glaucoma/pathology , Iron Chelating Agents/administration & dosage , Male , Mice , Mice, Inbred C57BL , Optic Nerve/drug effects , Oxidative Stress , Retinal Degeneration/etiology , Retinal Degeneration/pathology , Retinal Ganglion Cells/drug effects
20.
Mol Biol Rep ; 47(12): 9733-9738, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33249542

ABSTRACT

Akebia Saponin D (ASD), a triterpenoid saponin, was shown to have protective effects in certain neuronal cells. The purpose of the present study was to investigate the possibility of ASD to prevent tumor necrosis factor (TNF)-induced axonal loss and the ASD modulation of the biologic process of autophagy in optic nerves. Rats were given intravitreal administration of TNF, simultaneous administration of 2, 20, or 200 pmol ASD and TNF, or ASD alone. LC3-II and p62 expression, which is a marker of autophagic flux, and phosphorylated p38 (p-p38) expression in optic nerves were examined by immunoblot analysis. Morphometric analysis revealed a significant ameliorated effect of ASD against TNF-induced optic nerve damage. p62 was significantly increased in the optic nerve in TNF-treated eyes, but this increase was totally prevented by ASD. The ASD alone injection showed significant reduction of p62 levels compared with the PBS-treated control eyes. LC3-II was significantly increased by ASD treatment in the TNF-injected eyes. p-p38 was significantly increased in the optic nerve in TNF-treated eyes, but this increase was completely prevented by ASD. The protective effects of ASD may be associated with enhanced autophagy activation and inhibition of p-p38.


Subject(s)
Glaucoma/drug therapy , Nerve Degeneration/drug therapy , Neuroprotective Agents , Optic Nerve/drug effects , Saponins , Animals , Autophagy/drug effects , Axons/drug effects , Axons/pathology , Glaucoma/pathology , Male , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Optic Nerve/pathology , Rats , Rats, Wistar , Saponins/administration & dosage , Saponins/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL