Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 4881, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30890728

ABSTRACT

Antidepressants are commonly prescribed psychotropic substances for the symptomatic treatment of mood disorders. Their primary mechanism of action is the modulation of neurotransmission and the consequent accumulation of monoamines, such as serotonin and noradrenaline. However, antidepressants have additional molecular targets that, through multiple signaling cascades, may ultimately alter essential cellular processes. In this regard, it was previously demonstrated that clomipramine, a widely used FDA-approved tricyclic antidepressant, interferes with the autophagic flux and severely compromises the viability of tumorigenic cells upon cytotoxic stress. Consistent with this line of evidence, we report here that clomipramine undermines autophagosome formation and cargo degradation in primary dissociated neurons. A similar pattern was observed in the frontal cortex and liver of treated mice, as well as in the nematode Caenorhabditis elegans exposed to clomipramine. Together, our findings indicate that clomipramine may negatively regulate the autophagic flux in various tissues, with potential metabolic and functional implications for the homeostatic maintenance of differentiated cells.


Subject(s)
Affective Disorders, Psychotic/drug therapy , Antidepressive Agents, Tricyclic/pharmacology , Clomipramine/pharmacology , Neurons/drug effects , Affective Disorders, Psychotic/pathology , Animals , Antidepressive Agents, Tricyclic/adverse effects , Autophagy/drug effects , Caenorhabditis elegans/drug effects , Clomipramine/adverse effects , Disease Models, Animal , Liver/drug effects , Liver/metabolism , Mice , Neurons/metabolism , Norepinephrine/metabolism , Serotonin/metabolism , Signal Transduction/drug effects
2.
Cell Death Dis ; 9(10): 981, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30250019

ABSTRACT

Autophagy, the cellular process responsible for degradation and recycling of cytoplasmic components through the autophagosomal-lysosomal pathway, is fundamental for neuronal homeostasis and its deregulation has been identified as a hallmark of neurodegeneration. Retinal hypoxic-ischemic events occur in several sight-treating disorders, such as central retinal artery occlusion, diabetic retinopathy, and glaucoma, leading to degeneration and loss of retinal ganglion cells. Here we analyzed the autophagic response in the retinas of mice subjected to ischemia induced by transient elevation of intraocular pressure, reporting a biphasic and reperfusion time-dependent modulation of the process. Ischemic insult triggered in the retina an acute induction of autophagy that lasted during the first hours of reperfusion. This early upregulation of the autophagic flux limited RGC death, as demonstrated by the increased neuronal loss observed in mice with genetic impairment of basal autophagy owing to heterozygous ablation of the autophagy-positive modulator Ambra1 (Ambra1+/gt). Upregulation of autophagy was exhausted 24 h after the ischemic event and reduced autophagosomal turnover was associated with build up of the autophagic substrate SQSTM-1/p62, decreased ATG12-ATG5 conjugate, ATG4 and BECN1/Beclin1 expression. Animal fasting or subchronic systemic treatment with rapamycin sustained and prolonged autophagy activation and improved RGC survival, providing proof of principle for autophagy induction as a potential therapeutic strategy in retinal neurodegenerative conditions associated with hypoxic/ischemic stresses.


Subject(s)
Autophagy/drug effects , Autophagy/physiology , Cell Survival/drug effects , Fasting/metabolism , Reperfusion Injury/metabolism , Retinal Ganglion Cells/metabolism , Sirolimus/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Adenylate Kinase/metabolism , Animals , Autophagosomes/metabolism , Autophagy-Related Protein 12/metabolism , Autophagy-Related Protein 5/metabolism , Beclin-1/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Retina/metabolism , Sequestosome-1 Protein/metabolism , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases/metabolism
3.
Mol Vis ; 23: 911-921, 2017.
Article in English | MEDLINE | ID: mdl-29296071

ABSTRACT

Purpose: Retinal ischemic phenomena occur in several ocular diseases that share the degeneration and death of retinal ganglion cells (RGCs) as the final event. We tested the neuroprotective effect of azithromycin, a widely used semisynthetic macrolide antibiotic endowed with anti-inflammatory and immunomodulatory properties, in a model of retinal ischemic injury induced by transient elevation of intraocular pressure in the rat. Methods: Retinal ischemia was induced in adult rats with transient elevation of intraocular pressure. RGCs were retrogradely labeled with Fluoro-Gold, and survival was assessed following a single dose of azithromycin given systemically at the end of the ischemia. The expression of death-associated proteins and extracellular signal-regulated kinase (ERK) activation was studied with western blotting. Expression and activity of matrix metalloproteinase-2 (MMP-2) and -9 were analyzed with gelatin zymography. Results: Acute post-injury administration of azithromycin significantly prevented RGC death. This effect was accompanied by reduced calpain activity and prevention of Bcl-2-associated death promoter (Bad) upregulation. The observed neuroprotection was associated with a significant inhibition of MMP-2/-9 gelatinolytic activity and ERK1/2 phosphorylation. Conclusions: Azithromycin provides neuroprotection by modifying the inflammatory state of the retina following ischemia/reperfusion injury suggesting potential for repurposing as a drug capable of limiting or preventing retinal neuronal damage.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Azithromycin/therapeutic use , Disease Models, Animal , Reperfusion Injury/prevention & control , Retinal Diseases/prevention & control , Retinal Ganglion Cells/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Calpain/metabolism , Cell Survival/drug effects , Cytoprotection , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neuroprotective Agents/therapeutic use , Phosphorylation , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Retinal Diseases/metabolism , Retinal Ganglion Cells/metabolism , bcl-Associated Death Protein/metabolism
4.
Mol Vis ; 21: 718-29, 2015.
Article in English | MEDLINE | ID: mdl-26167113

ABSTRACT

PURPOSE: Retinal ganglion cell (RGC) death is the final event leading to visual impairment in glaucoma; therefore, identification of neuroprotective strategies able to slow down or prevent the process is one of the main challenges for glaucoma research. The purpose of this study was to evaluate the neuroprotective potential of RGC death induced by the in vivo transient increase in intraocular pressure (IOP) of a combined treatment with forskolin, homotaurine, and L-carnosine. Forskolin (7beta-acetoxy-8, 13-epoxy-1a, 6ß, 9a-trihydroxy-labd-14-en-11-one) is an activator of adenylate cyclase that decreases IOP by reducing aqueous humor production and functions as a neuroprotector due to its neurotrophin-stimulating activity. Homotaurine is a natural aminosulfonate compound endowed with neuromodulatory effects, while the dipeptide L-carnosine is known for its antioxidant properties. METHODS: Retinal ischemia was induced in the right eye of adult male Wistar rats by acutely increasing the IOP. Forskolin, homotaurine, and L-carnosine were intravitreally injected and RGC survival evaluated following retrograde labeling with FluoroGold. Total and phosphorylated Akt and glycogen synthase kinase-3ß (GSK-3ß) protein levels, as well as calpain activity, were analyzed with western blot. Protein kinase A (PKA) was inhibited by intravitreal injection of H89. RESULTS: A synergic neuroprotective effect on RGC survival was observed following the combined treatment with forskolin, homotaurine, and L-carnosine compared to forskolin alone. The observed neuroprotection was associated with reduced calpain activity, upregulation of phosphoinositide 3-kinase (PI3K)/Akt pathway, and inhibition of GSK-3ß but was independent from PKA activation and distinct from the hypotensive effects of forskolin. CONCLUSIONS: A multidrug/multitarget approach, by interfering with several pathways involved in RGC degeneration, may be promising to achieve glaucoma neuroprotection.


Subject(s)
Ischemia/drug therapy , Neuroprotective Agents/administration & dosage , Retinal Ganglion Cells/drug effects , Retinal Vessels/drug effects , Animals , Carnosine/administration & dosage , Cell Survival/drug effects , Colforsin/administration & dosage , Disease Models, Animal , Drug Synergism , Glaucoma/drug therapy , Glaucoma/pathology , Glaucoma/physiopathology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Intraocular Pressure , Intravitreal Injections , Ischemia/pathology , Ischemia/physiopathology , Male , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/physiology , Retinal Vessels/pathology , Retinal Vessels/physiopathology , Signal Transduction/drug effects , Taurine/administration & dosage , Taurine/analogs & derivatives
5.
Neurochem Int ; 79: 44-56, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25452081

ABSTRACT

Second messenger cAMP and cGMP represent a key step in the action of dopamine that modulates directly or indirectly their synthesis. We aimed to verify whether levodopa-induced dyskinesias are associated with changes of the time course of levodopa/dopamine stimulated cAMP and cGMP levels, and/or with changes of their catabolism by phosphodiesterase activity in rats with experimental hemiparkinsonism. Microdialysis and tissue homogenates of the striatal tissues demonstrated that extracellular and intracellular cAMP/cGMP levels were lower in dyskinetic animals during the increasing phase of dyskinesias compared to eukinetic animals, but cAMP/cGMP levels increased in dyskinetic animals during the phase of decreasing and extinction of dyskinesias. Dyskinesias and the abnormal lowering of striatal cGMP and cAMP after levodopa were prevented by pretreatment with the multipotent drug amantadine, outlining the inverse relationship of cAMP/cGMP to dyskinesias. Moreover, dyskinetic animals showed higher striatal hydrolyzing cGMP-phosphodiesterase but not hydrolyzing cAMP-phosphodiesterase activity, suggesting that low cGMP but not cAMP levels could be due to increased catabolism. However, expressions of isozyme phosphodiesterase-1B and -10A highly and specifically located in the basal ganglia were not changed after levodopa in dyskinetic and eukinetic animals: accordingly, selective inhibitors of phosphodiesterase-1B and -10A were ineffective on levodopa dyskinesias. Therefore, the isozyme(s) expressing higher cGMP-phosphodiesterase activity in the striatum of dyskinetic animal should be determined. These observations suggest that dopamine-mediated processes of synthesis and/or degradation of cAMP/cGMP could be acutely impaired in levodopa dyskinesias, opening new ways to understanding physiopathology and treatment.


Subject(s)
Antiparkinson Agents/adverse effects , Caudate Nucleus/drug effects , Caudate Nucleus/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Dyskinesia, Drug-Induced/metabolism , Dyskinesia, Drug-Induced/physiopathology , Levodopa/adverse effects , Parkinsonian Disorders/drug therapy , Putamen/drug effects , Putamen/metabolism , Animals , Down-Regulation/drug effects , Male , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Rats , Rats, Wistar
6.
PLoS One ; 8(8): e69250, 2013.
Article in English | MEDLINE | ID: mdl-23936321

ABSTRACT

Excitotoxicity has been implicated in the retinal neuronal loss in several ocular pathologies including glaucoma. Dysfunction of Excitatory Amino Acid Transporters is often a key component of the cascade leading to excitotoxic cell death. In the retina, glutamate transport is mainly operated by the glial glutamate transporter GLAST and the neuronal transporter GLT-1. In this study we evaluated the expression of GLAST and GLT-1 in a rat model of acute glaucoma based on the transient increase of intraocular pressure (IOP) and characterized by high glutamate levels during the reperfusion that follows the ischemic event associated with raised IOP. No changes were reported in GLAST expression while, at neuronal level, a reduction of glutamate uptake and of transporter reversal-mediated glutamate release was observed in isolated retinal synaptosomes. This was accompanied by modulation of GLT-1 expression leading to the reduction of the canonical 65 kDa form and upregulation of a GLT-1-related 38 kDa protein. These results support a role for neuronal transporters in glutamate accumulation observed in the retina following an ischemic event and suggest the presence of a GLT-1 neuronal new alternative splice variant, induced in response to the detrimental stimulus.


Subject(s)
Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/metabolism , Ischemia/metabolism , Ischemia/pathology , Neurons/metabolism , Retina/pathology , Retina/physiopathology , Animals , Aspartic Acid/metabolism , Biological Transport , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/biosynthesis , Gene Expression Regulation , Intraocular Pressure , Male , Neurons/pathology , Rats , Rats, Wistar , Retina/metabolism , Synaptosomes/metabolism
7.
Curr Eye Res ; 38(4): 451-63, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23373736

ABSTRACT

PURPOSE: Adipose-derived stem cells (ADSC) are multipotent, safe, non-immunogenic and can differentiate into functional keratocytes in situ. The topical use of ADSC derived from human processed lipoaspirate was investigated for treating injured rat cornea. METHODS: A total of 19 rats were used. Six animals initially underwent corneal lesion experiments with 0.5 N NaOH (right eye) and 0.2 N (left). The 0.2 NaOH protocol was then used in 13 rats. All 26 eyes of 13 rats eyes received topical azythromycin bid for 3 d and divided into five treatment groups (n = 5 eyes/group), which included: control, stem cells, serum, stem + serum and adipose (raw human lipoaspirate). The four treatment groups received topical treatment three times daily for 3 d. Stem cells were isolated and harvested from human lipoaspirate. Topical eye drops were prepared daily with 1 × 10(5) cells/treatment. Fluorescein positive defect area and light microscope assessment was performed at 20, 28, 45, 50 and 74 h. Animals were sacrificed at 74 h for histological evaluation. Data were statistically analyzed for differences amongst groups. RESULTS: The stem cell-treated eyes had significantly smaller epithelial defects at each time point compared to control- and adipose-treated eyes (p < 0.05). This group showed slightly better epithelium healing than the serum and combined group, yet not significantly different. Histology showed that stem cell-treated corneas had complete re-epithelization, with less inflammatory cells and limited fibroblast activation structure compared with the control eyes. CONCLUSIONS: Our preliminary results show that topical treatment with ADSC seems to improve corneal wound healing.


Subject(s)
Adipose Tissue/cytology , Burns, Chemical/surgery , Epithelium, Corneal/surgery , Eye Burns/surgery , Stem Cell Transplantation/methods , Animals , Burns, Chemical/pathology , Burns, Chemical/physiopathology , Disease Models, Animal , Epithelium, Corneal/pathology , Epithelium, Corneal/physiology , Eye Burns/pathology , Eye Burns/physiopathology , Fluorescent Dyes , Humans , Male , Pilot Projects , Rats , Rats, Wistar , Staining and Labeling , Statistics, Nonparametric , Transplantation, Heterologous , Wound Healing/physiology
8.
Prog Brain Res ; 173: 575-82, 2008.
Article in English | MEDLINE | ID: mdl-18929135

ABSTRACT

Glaucoma is a worldwide leading cause of irreversible vision loss characterized by progressive death of retinal ganglion cells (RGCs). In the course of glaucoma, RGC death may be the consequence of energy impairment that triggers secondary excitotoxicity and free radical generation. There is substantial evidence also that a number of free radical scavengers and/or agents that improve mitochondrial function may be useful as therapies to ameliorate cell death in various neurological disorders including glaucoma. Coenzyme Q10 (CoQ10), an essential cofactor of the electron transport chain, has been reported to afford neuroprotection in neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases, and its protective effect has been attributed in part to its free radical scavenger ability and to a specific regulation of the mitochondrial permeability transition pore. Using an established animal model of transient retinal ischemia, we have conclusively identified a role for abnormal elevation of extracellular glutamate in the mechanisms underlying RGC death that occurs, at least in part, via activation of the apoptotic program. Under these experimental conditions, N-methyl-D-aspartate (NMDA) and non-NMDA subtype of glutamate receptor antagonists, nitric oxide synthase inhibitors, and CoQ10 afford retinal protection supporting an important role for excitotoxicity in the mechanisms underlying RGC death.


Subject(s)
Glaucoma/drug therapy , Neuroprotective Agents , Retina/drug effects , Ubiquinone/analogs & derivatives , Animals , Calpain/metabolism , Cell Death/physiology , Glaucoma/pathology , Glutamic Acid/metabolism , Humans , Intraocular Pressure/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress , Rats , Reperfusion Injury/prevention & control , Retina/cytology , Retina/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Ubiquinone/pharmacology , Ubiquinone/therapeutic use
9.
Prog Brain Res ; 173: 583-90, 2008.
Article in English | MEDLINE | ID: mdl-18929136

ABSTRACT

Glaucoma, is a progressive optic neuropathy often associated with increased intraocular pressure (IOP) and characterized by progressive death of retinal ganglion cells (RGCs). High acute rise of IOP is a model for retinal ischemia and may represent a model of acute angle closure glaucoma. Here we have used this experimental model in combination with a neurochemical and neuropathological approach to gain more insight in the neuroprotective profile of 17beta-estradiol (E2), a steroid hormone, which has been shown to increase the viability, survival, and differentiation of primary neuronal cultures from different brain areas including amygdala, hypothalamus, and neocortex. Our data demonstrate that systemic administration of E2 significantly reduces RGC loss induced by high IOP in rat. In addition, pretreatment with E2, 30 min before ischemia, minimizes the elevation of glutamate observed during the reperfusion period. These effects seem to be in part mediated by the activation of the estrogen receptor, since a pretreatment with ICI 182-780, a specific estrogen receptor antagonist, partially counteracts the neuroprotection afforded by the estrogen.


Subject(s)
Estradiol/pharmacology , Estrogens/pharmacology , Intraocular Pressure/drug effects , Retinal Ganglion Cells , Animals , Cells, Cultured , Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Fulvestrant , Glaucoma/pathology , Glaucoma/physiopathology , Glutamic Acid/metabolism , Male , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Rats , Rats, Wistar , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology
10.
Int Rev Neurobiol ; 82: 397-406, 2007.
Article in English | MEDLINE | ID: mdl-17678974

ABSTRACT

Recent studies support a role for excitotoxicity in the development of retinal ganglion cell (RGC) damage in subjects suffering from glaucoma. Coenzyme Q10 (CoQ10), an essential cofactor of the electron transport chain, has been reported to afford neuroprotection, preventing the formation of the mitochondrial permeability transition pore. Using an established animal model of retinal ischemia/reperfusion here, we show that synaptic glutamate increases at 130min from beginning of reperfusion and delayed apoptosis in the RGC layer is seen at 24h. Intraocular administration of CoQ10 minimizes glutamate increase and affords neuroprotection, suggesting that oxidative stress and energy failure might be implicated in the mechanisms of RGC death.


Subject(s)
Intraocular Pressure/physiology , Ischemia/pathology , Retinal Diseases/prevention & control , Ubiquinone/analogs & derivatives , Administration, Topical , Animals , Coenzymes/administration & dosage , Coenzymes/therapeutic use , Glutamic Acid/toxicity , In Situ Nick-End Labeling , Male , Microdialysis , Rats , Rats, Wistar , Retinal Diseases/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Ubiquinone/administration & dosage , Ubiquinone/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...