Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Polymers (Basel) ; 16(8)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38675037

ABSTRACT

Corneal diseases represent a significant global health challenge, often resulting in blindness, for which penetrating keratoplasty is the clinical gold standard. However, in cases involving compromised ocular surfaces or graft failure, osteo-odonto keratoprosthesis (OOKP) emerges as a vital yet costly and complex alternative. Thus, there is an urgent need to introduce soft biomaterials that mimic the corneal tissue, considering its translation's physicochemical, biological, and economic costs. This study introduces a cross-linked mixture of economically viable biomaterials, including gelatin, chitosan, and poly-D-lysine, that mimic corneal properties. The physicochemical evaluation of certain mixtures, specifically gelatin, chitosan, and poly-D-lysine cross-linked with 0.10% glutaraldehyde, demonstrates that properties such as swelling, optical transmittance, and thermal degradation are comparable to those of native corneas. Additionally, constructs fabricated with poly-D-lysine exhibit good cytocompatibility with fibroblasts at 72 h. These findings suggest that low-cost biopolymers, particularly those incorporating poly-D-lysine, mimic specific corneal characteristics and have the potential to foster fibroblast survival. While further studies are required to reach a final corneal-mimicking solution, this study contributes to positioning low-cost reagents as possible alternatives to develop biomaterials with physicochemical properties like those of the human cornea.

2.
Discov Nano ; 18(1): 129, 2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37847425

ABSTRACT

Carbon dots (CDs) have been quickly extended for nanomedicine uses because of their multiple applications, such as bioimaging, sensors, and drug delivery. However, the interest in increasing their photoluminescence properties is not always accompanied by cytocompatibility. Thus, a knowledge gap exists regarding their interactions with biological systems linked to the selected formulations and synthesis methods. In this work, we have developed carbon dots (CDs) based on poly (ethylene imine) (PEI) and chitosan (CS) by using microwave irradiation, hydrothermal synthesis, and a combination of both, and further characterized them by physicochemical and biological means. Our results indicate that synthesized CDs have sizes between 1 and 5 nm, a high presence of amine groups on the surface, and increased positive ζ potential values. Further, it is established that the choice and use of different synthesis procedures can contribute to a different answer to the CDs regarding their optical and biological properties. In this regard, PEI-only CDs showed the longest photoluminescent emission lifetime, non-hemolytic activity, and high toxicity against fibroblast. On the other hand, CS-only CDs have higher PL emission, non-cytotoxicity associated with fibroblast, and high hemolytic activity. Interestingly, their combination using the proposed methodologies allow a synergic effect in their CDs properties. Therefore, this work contributes to developing and characterizing CD formulations based on PEI and CS and better understanding the CD's properties and biological interaction.

3.
Front Mol Neurosci ; 16: 1241222, 2023.
Article in English | MEDLINE | ID: mdl-37736113

ABSTRACT

Dominant optic atrophy (DOA) is mainly caused by OPA1 mutations and is characterized by the degeneration of retinal ganglion cells (RGCs), whose axons form the optic nerve. The penetrance of DOA is incomplete and the disease is marked by highly variable expressivity, ranging from asymptomatic patients to some who are totally blind or who suffer from multisystemic effects. No clear genotype-phenotype correlation has been established to date. Taken together, these observations point toward the existence of modifying genetic and/or environmental factors that modulate disease severity. Here, we investigated the influence of genetic background on DOA expressivity by switching the previously described DOA mouse model bearing the c.1065 + 5G → A Opa1 mutation from mixed C3H; C57BL/6 J to a pure C57BL/6 J background. We no longer observed retinal and optic nerve abnormalities; the findings indicated no degeneration, but rather a sex-dependent negative effect on RGC connectivity. This highlights the fact that RGC synaptic alteration might precede neuronal death, as has been proposed in other neurodegenerative diseases, providing new clinical considerations for early diagnosis as well as a new therapeutic window for DOA. Furthermore, our results demonstrate the importance of secondary genetic factors in the variability of DOA expressivity and offer a model for screening for aggravating environmental and genetic factors.

4.
Aging Cell ; 22(5): e13814, 2023 05.
Article in English | MEDLINE | ID: mdl-36973898

ABSTRACT

Age is the main risk factor for the development of neurodegenerative diseases. In the aged brain, axonal degeneration is an early pathological event, preceding neuronal dysfunction, and cognitive disabilities in humans, primates, rodents, and invertebrates. Necroptosis mediates degeneration of injured axons, but whether necroptosis triggers neurodegeneration and cognitive impairment along aging is unknown. Here, we show that the loss of the necroptotic effector Mlkl was sufficient to delay age-associated axonal degeneration and neuroinflammation, protecting against decreased synaptic transmission and memory decline in aged mice. Moreover, short-term pharmacologic inhibition of necroptosis targeting RIPK3 in aged mice, reverted structural and functional hippocampal impairment, both at the electrophysiological and behavioral level. Finally, a quantitative proteomic analysis revealed that necroptosis inhibition leads to an overall improvement of the aged hippocampal proteome, including a subclass of molecular biofunctions associated with brain rejuvenation, such as long-term potentiation and synaptic plasticity. Our results demonstrate that necroptosis contributes to age-dependent brain degeneration, disturbing hippocampal neuronal connectivity, and cognitive function. Therefore, necroptosis inhibition constitutes a potential geroprotective strategy to treat age-related disabilities associated with memory impairment and cognitive decline.


Subject(s)
Necroptosis , Neurodegenerative Diseases , Humans , Mice , Animals , Aged , Proteomics , Rejuvenation , Aging/physiology , Brain , Memory Disorders
6.
Sci Rep ; 11(1): 7395, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33795747

ABSTRACT

Wnt signaling plays a key role in neurodevelopment and neuronal maturation. Specifically, Wnt5a stimulates postsynaptic assemblies, increases glutamatergic neurotransmission and, through calcium signaling, generates nitric oxide (NO). Trying to unveil the molecular pathway triggering these postsynaptic effects, we found that Wnt5a treatment induces a time-dependent increases in the length of the postsynaptic density (PSD), elicits novel synaptic contacts and facilitates F-actin flow both in in vitro and ex vivo models. These effects were partially abolished by the inhibition of the Heme-regulated eukaryotic initiation factor 2α (HRI) kinase, a kinase which phosphorylates the initiation translational factor eIF2α. When phosphorylated, eIF2α normally avoids the translation of proteins not needed during stress conditions, in order to avoid unnecessary energetic expenses. However, phosphorylated eIF2α promotes the translation of some proteins with more than one open reading frame in its 5' untranslated region. One of these proteins targeted by Wnt-HRI-eIF2α mediated translation is the GluN2B subunit of the NMDA receptor. The identified increase in GluN2B expression correlated with increased NMDA receptor function. Considering that NMDA receptors are crucial for excitatory synaptic transmission, the molecular pathway described here contributes to the understanding of the fast and plastic translational mechanisms activated during learning and memory processes.


Subject(s)
Hippocampus/growth & development , Protein Serine-Threonine Kinases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Wnt-5a Protein/metabolism , 5' Untranslated Regions , Actins/metabolism , Animals , Culture Media, Conditioned , Gene Expression Regulation , Hippocampus/metabolism , Learning , Male , Memory , Mice , Mice, Inbred C57BL , Neuronal Plasticity , Nitric Oxide/metabolism , Open Reading Frames , Phosphorylation , RNA, Small Interfering/metabolism , Signal Transduction , Synapses/metabolism , Synaptosomes/metabolism
7.
Pharmaceuticals (Basel) ; 13(2)2020 Feb 02.
Article in English | MEDLINE | ID: mdl-32024240

ABSTRACT

Alzheimer's Disease (AD) is the primary cause of dementia among the elderly population. Elevated plasma levels of homocysteine (HCy), an amino acid derived from methionine metabolism, are considered a risk factor and biomarker of AD and other types of dementia. An increase in HCy is mostly a consequence of high methionine and/or low vitamin B intake in the diet. Here, we studied the effects of physiological and pathophysiological HCy concentrations on oxidative stress, synaptic protein levels, and synaptic activity in mice hippocampal slices. We also studied the in vitro effects of HCy on the aggregation kinetics of Aß40. We found that physiological cerebrospinal concentrations of HCy (0.5 µM) induce an increase in synaptic proteins, whereas higher doses of HCy (30-100 µM) decrease their levels, thereby increasing oxidative stress and causing excitatory transmission hyperactivity, which are all considered to be neurotoxic effects. We also observed that normal cerebrospinal concentrations of HCy slow the aggregation kinetic of Aß40, whereas high concentrations accelerate its aggregation. Finally, we studied the effects of HCy and HCy + Aß42 over long-term potentiation. Altogether, by studying an ample range of effects under different HCy concentrations, we report, for the first time, that HCy can exert beneficial or toxic effects over neurons, evidencing a hormetic-like effect. Therefore, we further encourage the use of HCy as a biomarker and modifiable risk factor with therapeutic use against AD and other types of dementia.

9.
Sci Rep ; 9(1): 6107, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988455

ABSTRACT

Pathogenic variants of OPA1, which encodes a dynamin GTPase involved in mitochondrial fusion, are responsible for a spectrum of neurological disorders sharing optic nerve atrophy and visual impairment. To gain insight on OPA1 neuronal specificity, we performed targeted metabolomics on rat cortical neurons with OPA1 expression inhibited by RNA interference. Of the 103 metabolites accurately measured, univariate analysis including the Benjamini-Hochberg correction revealed 6 significantly different metabolites in OPA1 down-regulated neurons, with aspartate being the most significant (p < 0.001). Supervised multivariate analysis by OPLS-DA yielded a model with good predictive capability (Q2cum = 0.65) and a low risk of over-fitting (permQ2 = -0.16, CV-ANOVA p-value 0.036). Amongst the 46 metabolites contributing the most to the metabolic signature were aspartate, glutamate and threonine, which all decreased in OPA1 down-regulated neurons, and lysine, 4 sphingomyelins, 4 lysophosphatidylcholines and 32 phosphatidylcholines which were increased. The phospholipid signature may reflect intracellular membrane remodeling due to loss of mitochondrial fusion and/or lipid droplet accumulation. Aspartate and glutamate deficiency, also found in the plasma of OPA1 patients, is likely the consequence of respiratory chain deficiency, whereas the glutamate decrease could contribute to the synaptic dysfunction that we previously identified in this model.


Subject(s)
Cerebral Cortex/pathology , GTP Phosphohydrolases/deficiency , Neurons/pathology , Optic Atrophy, Autosomal Dominant/pathology , Animals , Aspartic Acid/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Down-Regulation , Embryo, Mammalian , Female , GTP Phosphohydrolases/genetics , Glutamic Acid/metabolism , Humans , Metabolomics , Optic Atrophy, Autosomal Dominant/genetics , Phospholipids/metabolism , Primary Cell Culture , RNA, Small Interfering/metabolism , Rats
10.
J Neurosci ; 39(20): 3832-3844, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30850513

ABSTRACT

Axonal degeneration, which contributes to functional impairment in several disorders of the nervous system, is an important target for neuroprotection. Several individual factors and subcellular events have been implicated in axonal degeneration, but researchers have so far been unable to identify an integrative signaling pathway activating this self-destructive process. Through pharmacological and genetic approaches, we tested whether necroptosis, a regulated cell-death mechanism implicated in the pathogenesis of several neurodegenerative diseases, is involved in axonal degeneration. Pharmacological inhibition of the necroptotic kinase RIPK1 using necrostatin-1 strongly delayed axonal degeneration in the peripheral nervous system and CNS of wild-type mice of either sex and protected in vitro sensory axons from degeneration after mechanical and toxic insults. These effects were also observed after genetic knock-down of RIPK3, a second key regulator of necroptosis, and the downstream effector MLKL (Mixed Lineage Kinase Domain-Like). RIPK1 inhibition prevented mitochondrial fragmentation in vitro and in vivo, a typical feature of necrotic death, and inhibition of mitochondrial fission by Mdivi also resulted in reduced axonal loss in damaged nerves. Furthermore, electrophysiological analysis demonstrated that inhibition of necroptosis delays not only the morphological degeneration of axons, but also the loss of their electrophysiological function after nerve injury. Activation of the necroptotic pathway early during injury-induced axonal degeneration was made evident by increased phosphorylation of the downstream effector MLKL. Our results demonstrate that axonal degeneration proceeds by necroptosis, thus defining a novel mechanistic framework in the axonal degenerative cascade for therapeutic interventions in a wide variety of conditions that lead to neuronal loss and functional impairment.SIGNIFICANCE STATEMENT We show that axonal degeneration triggered by diverse stimuli is mediated by the activation of the necroptotic programmed cell-death program by a cell-autonomous mechanism. This work represents a critical advance for the field since it identifies a defined degenerative pathway involved in axonal degeneration in both the peripheral nervous system and the CNS, a process that has been proposed as an early event in several neurodegenerative conditions and a major contributor to neuronal death. The identification of necroptosis as a key mechanism for axonal degeneration is an important step toward the development of novel therapeutic strategies for nervous-system disorders, particularly those related to chemotherapy-induced peripheral neuropathies or CNS diseases in which axonal degeneration is a common factor.


Subject(s)
Axons/physiology , Mitochondria/physiology , Necroptosis/physiology , Nerve Degeneration/physiopathology , Animals , Cells, Cultured , Dynamins/physiology , Female , Gene Knockdown Techniques , Male , Mice, Inbred C57BL , Optic Nerve Injuries/physiopathology , Protein Kinases/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Wallerian Degeneration/physiopathology
11.
Neurotox Res ; 36(2): 257-267, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30215161

ABSTRACT

Generation of new neurons is a tightly regulated process that involves several intrinsic and extrinsic factors. Among them, a metabolic switch from glycolysis to oxidative phosphorylation, together with mitochondrial remodeling, has emerged as crucial actors of neurogenesis. However, although accumulating data raise the importance of mitochondrial morphology and function in neural stem cell proliferation and differentiation during development, information regarding the contribution of mitochondria to adult neurogenesis processes remains limited. In the present review, we discuss recent evidence covering the importance of mitochondrial morphology, function, and energy metabolism in the regulation of neuronal development and adult neurogenesis, and their impact on memory processes.


Subject(s)
Mitochondria/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Adult , Animals , Cell Differentiation/physiology , Humans
12.
Neurobiol Dis ; 102: 113-124, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28286181

ABSTRACT

Adult hippocampal neurogenesis is strongly impaired in Alzheimer's disease (AD). In several mouse models of AD, it was shown that adult-born neurons exhibit reduced survival and altered synaptic integration due to a severe lack of dendritic spines. In the present work, using the APPxPS1 mouse model of AD, we reveal that this reduced number of spines is concomitant of a marked deficit in their neuronal mitochondrial content. Remarkably, we show that targeting the overexpression of the pro-neural transcription factor Neurod1 into APPxPS1 adult-born neurons restores not only their dendritic spine density, but also their mitochondrial content and the proportion of spines associated with mitochondria. Using primary neurons, a bona fide model of neuronal maturation, we identified that increases of mitochondrial respiration accompany the stimulating effect of Neurod1 overexpression on dendritic growth and spine formation. Reciprocally, pharmacologically impairing mitochondria prevented Neurod1-dependent trophic effects. Thus, since overexpression of Neurod1 into new neurons of APPxPS1 mice rescues spatial memory, our present data suggest that manipulating the mitochondrial system of adult-born hippocampal neurons provides neuronal plasticity to the AD brain. These findings open new avenues for far-reaching therapeutic implications towards neurodegenerative diseases associated with cognitive impairment.


Subject(s)
Alzheimer Disease/metabolism , Dendritic Spines/metabolism , Mitochondria/metabolism , Neurogenesis/physiology , Alzheimer Disease/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Dendritic Spines/pathology , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice, Transgenic , Mitochondria/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Organelle Biogenesis , Random Allocation , Rats, Wistar
13.
PLoS One ; 12(1): e0168840, 2017.
Article in English | MEDLINE | ID: mdl-28060833

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder mainly known for synaptic impairment and neuronal cell loss, affecting memory processes. Beside these damages, mitochondria have been implicated in the pathogenesis of AD through the induction of the mitochondrial permeability transition pore (mPTP). The mPTP is a non-selective pore that is formed under apoptotic conditions, disturbing mitochondrial structure and thus, neuronal viability. In AD, Aß oligomers (Aßos) favor the opening of the pore, activating mitochondria-dependent neuronal cell death cascades. The Wnt signaling activated through the ligand Wnt3a has been described as a neuroprotective signaling pathway against amyloid-ß (Aß) peptide toxicity in AD. However, the mechanisms by which Wnt signaling prevents Aßos-induced neuronal cell death are unclear. We proposed here to study whether Wnt signaling protects neurons earlier than the late damages in the progression of the disease, through the preservation of the mitochondrial structure by the mPTP inhibition. To study specific events related to mitochondrial permeabilization we performed live-cell imaging from primary rat hippocampal neurons, and electron microscopy to analyze the mitochondrial morphology and structure. We report here that Wnt3a prevents an Aßos-induced cascade of mitochondrial events that leads to neuronal cell death. This cascade involves (a) mPTP opening, (b) mitochondrial swelling, (c) mitochondrial membrane potential loss and (d) cytochrome c release, thus leading to neuronal cell death. Furthermore, our results suggest that the activation of the Wnt signaling prevents mPTP opening by two possible mechanisms, which involve the inhibition of mitochondrial GSK-3ß and/or the modulation of mitochondrial hexokinase II levels and activity. This study suggests a possible new approach for the treatment of AD from a mitochondrial perspective, and will also open new lines of study in the field of Wnt signaling in neuroprotection.


Subject(s)
Amyloid beta-Peptides/metabolism , Hippocampus/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Neurons/metabolism , Wnt Signaling Pathway , Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Animals , Cells, Cultured , Female , Glycogen Synthase Kinase 3 beta/metabolism , Hexokinase/metabolism , Hippocampus/cytology , Hippocampus/ultrastructure , Mitochondrial Membranes/ultrastructure , Mitochondrial Swelling , Neurons/ultrastructure , Permeability , Phosphorylation , Pregnancy , Rats , Rats, Sprague-Dawley , Wnt3A Protein/metabolism
14.
Mol Neurodegener ; 10: 62, 2015 Nov 21.
Article in English | MEDLINE | ID: mdl-26590557

ABSTRACT

BACKGROUND: L-methionine, the principal sulfur-containing amino acid in proteins, plays critical roles in cell physiology as an antioxidant and in the breakdown of fats and heavy metals. Previous studies suggesting the use of L-methionine as a treatment for depression and other diseases indicate that it might also improve memory and propose a role in brain function. However, some evidence indicates that an excess of methionine can be harmful and can increase the risk of developing Type-2 diabetes, heart diseases, certain types of cancer, brain alterations such as schizophrenia, and memory impairment. RESULTS: Here, we report the effects of an L-methionine-enriched diet in wild-type mice and emphasize changes in brain structure and function. The animals in our study presented 1) higher levels of phosphorylated tau protein, 2) increased levels of amyloid-ß (Aß)-peptides, including the formation of Aß oligomers, 3) increased levels of inflammatory response,4) increased oxidative stress, 5) decreased level of synaptic proteins, and 6) memory impairment and loss. We also observed dysfunction of the Wnt signaling pathway. CONCLUSION: Taken together, the results of our study indicate that an L-methionine-enriched diet causes neurotoxic effects in vivo and might contribute to the appearance of Alzheimer's-like neurodegeneration.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Behavior, Animal , Brain/metabolism , Neurons/metabolism , Wnt Signaling Pathway , tau Proteins/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Behavior, Animal/physiology , Female , Methionine/metabolism , Mice, Inbred C57BL , Oxidative Stress/physiology , Phosphorylation , Wnt Signaling Pathway/physiology
15.
Front Cell Neurosci ; 9: 166, 2015.
Article in English | MEDLINE | ID: mdl-25999816

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disorder and is characterized by progressive memory loss and cognitive decline. One of the hallmarks of AD is the overproduction of amyloid-beta aggregates that range from the toxic soluble oligomer (Aßo) form to extracellular accumulations in the brain. Growing evidence indicates that mitochondrial dysfunction is a common feature of neurodegenerative diseases and is observed at an early stage in the pathogenesis of AD. Reports indicate that mitochondrial structure and function are affected by Aßo and can trigger neuronal cell death. Mitochondria are highly dynamic organelles, and the balance between their fusion and fission processes is essential for neuronal function. Interestingly, in AD, the process known as "mitochondrial dynamics" is also impaired by Aßo. On the other hand, the activation of the Wnt signaling pathway has an essential role in synaptic maintenance and neuronal functions, and its deregulation has also been implicated in AD. We have demonstrated that canonical Wnt signaling, through the Wnt3a ligand, prevents the permeabilization of mitochondrial membranes through the inhibition of the mitochondrial permeability transition pore (mPTP), induced by Aßo. In addition, we showed that non-canonical Wnt signaling, through the Wnt5a ligand, protects mitochondria from fission-fusion alterations in AD. These results suggest new approaches by which different Wnt signaling pathways protect neurons in AD, and support the idea that mitochondria have become potential therapeutic targets for the treatment of neurodegenerative disorders. Here we discuss the neuroprotective role of the canonical and non-canonical Wnt signaling pathways in AD and their differential modulation of mitochondrial processes, associated with mitochondrial dysfunction and neurodegeneration.

16.
Exp Neurol ; 264: 14-25, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25450465

ABSTRACT

Amyloid-ß (Aß) oligomers are a key factor in Alzheimer's disease (AD)-associated synaptic dysfunction. Aß oligomers block the induction of hippocampal long-term potentiation (LTP) in rodents. The activation of Wnt signaling prevents Aß oligomer-induced neurotoxic effects. The compound WASP-1 (Wnt-activating small molecule potentiator-1), has been described as a synergist of the ligand Wnt-3a, enhancing the activation of Wnt/ß-catenin signaling. Herein, we report that WASP-1 administration successfully rescued Aß-induced synaptic impairments both in vitro and in vivo. The activation of canonical Wnt/ß-catenin signaling by WASP-1 increased synaptic transmission and rescued hippocampal LTP impairments induced by Aß oligomers. Additionally, intra-hippocampal administration of WASP-1 to the double transgenic APPswe/PS1dE9 mouse model of AD prevented synaptic protein loss and reduced tau phosphorylation levels. Moreover, we found that WASP-1 blocked Aß aggregation in vitro and reduced pathological tau phosphorylation in vivo. These results indicate that targeting canonical Wnt signaling with WASP-1 could have value for treating AD.


Subject(s)
CCN Intercellular Signaling Proteins/therapeutic use , Hippocampus/drug effects , Neurons/drug effects , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/pathology , Proto-Oncogene Proteins/therapeutic use , Synapses/drug effects , Amyloid beta-Peptides/toxicity , Amyloid beta-Protein Precursor/genetics , Animals , Cells, Cultured , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hippocampus/pathology , Hippocampus/physiology , Humans , Male , Mice , Mice, Transgenic , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Presenilin-1/genetics , Rats , Signal Transduction/drug effects , Signal Transduction/genetics , Synapses/genetics , Synapses/physiology , Synapses/ultrastructure , Time Factors
17.
Methods Mol Biol ; 1254: 87-97, 2015.
Article in English | MEDLINE | ID: mdl-25431059

ABSTRACT

Maintenance of mitochondrial membrane integrity is essential for mitochondrial function and neuronal viability. Apoptotic stimulus or calcium overload leads to mitochondrial permeability transition pore (mPTP ) opening and induces mitochondrial swelling, a common feature of mitochondrial membrane permeabilization. The first phenomenon can be evaluated in cells loaded with the dye calcein -AM quenched by cobalt, and mitochondrial swelling can be detected by electron microscopy through the analysis of mitochondrial membrane integrity. Here, we describe a live cell imaging assay to detect mitochondrial permeability transition and the development of a detailed analysis of morphological and ultrastructural changes that mitochondria undergo during this process.


Subject(s)
Microscopy, Electron , Mitochondrial Membrane Transport Proteins/ultrastructure , Mitochondrial Membranes/ultrastructure , Neurons/ultrastructure , Calcium/metabolism , Mitochondria/ultrastructure , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling , Molecular Biology/methods , Neurons/metabolism , Permeability
18.
Antioxid Redox Signal ; 22(1): 48-62, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-24988428

ABSTRACT

AIMS: To examine the role of the enzyme methionine sulfoxide reductase A-1 (MSRA-1) in amyloid-ß peptide (Aß)-peptide aggregation and toxicity in vivo, using a Caenorhabditis elegans model of the human amyloidogenic disease inclusion body myositis. RESULTS: MSRA-1 specifically reduces oxidized methionines in proteins. Therefore, a deletion of the msra-1 gene was introduced into transgenic C. elegans worms that express the Aß-peptide in muscle cells to prevent the reduction of oxidized methionines in proteins. In a constitutive transgenic Aß strain that lacks MSRA-1, the number of amyloid aggregates decreases while the number of oligomeric Aß species increases. These results correlate with enhanced synaptic dysfunction and mislocalization of the nicotinic acetylcholine receptor ACR-16 at the neuromuscular junction (NMJ). INNOVATION: This approach aims at modulating the oxidation of Aß in vivo indirectly by dismantling the methionine sulfoxide repair system. The evidence presented here shows that the absence of MSRA-1 influences Aß aggregation and aggravates locomotor behavior and NMJ dysfunction. The results suggest that therapies which boost the activity of the Msr system could have a beneficial effect in managing amyloidogenic pathologies. CONCLUSION: The absence of MSRA-1 modulates Aß-peptide aggregation and increments its deleterious effects in vivo.


Subject(s)
Methionine Sulfoxide Reductases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Animals, Genetically Modified , Blotting, Western , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Immunoprecipitation , Locomotion/physiology , Methionine , Oxidation-Reduction , Receptors, Nicotinic/metabolism
19.
J Biol Chem ; 289(52): 36179-93, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25336659

ABSTRACT

The Wnt signaling pathway plays an important role in developmental processes, including embryonic patterning, cell specification, and cell polarity. Wnt components participate in the development of the central nervous system, and growing evidence indicates that this pathway also regulates the function of the adult nervous system. In this study, we report that Wnt-5a, a noncanonical Wnt ligand, is a potent activator of mitochondrial dynamics and induces acute fission and fusion events in the mitochondria of rat hippocampal neurons. The effect of Wnt-5a was inhibited in the presence of sFRP, a Wnt scavenger. Similarly, the canonical Wnt-3a ligand had no effect on mitochondrial fission-fusion events, suggesting that this effect is specific for Wnt-5a alone. We also show that the Wnt-5a effects on mitochondrial dynamics occur with an increase in both intracellular and mitochondrial calcium (Ca(2+)), which was correlated with an increased phosphorylation of Drp1(Ser-616) and a decrease of Ser-637 phosphorylation, both indicators of mitochondrial dynamics. Electron microscope analysis of hippocampal tissues in the CA1 region showed an increase in the number of mitochondria present in the postsynaptic region, and this finding correlated with a change in mitochondrial morphology. We conclude that Wnt-5a/Ca(2+) signaling regulates the mitochondrial fission-fusion process in hippocampal neurons, a feature that might help to further understand the role of Wnt-related pathologies, including neurodegenerative diseases associated with mitochondrial dysfunction, and represents a potentially important link between impaired metabolic function and degenerative disorders.


Subject(s)
Mitochondrial Dynamics , Wnt Proteins/physiology , Animals , CA1 Region, Hippocampal/cytology , Calcium Signaling , Cells, Cultured , Dynamins/metabolism , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Mitochondria/physiology , Mitochondria/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Protein Processing, Post-Translational , Protein Transport , Rats, Sprague-Dawley , Wnt-5a Protein
20.
Neuromolecular Med ; 15(3): 549-69, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23842742

ABSTRACT

An emerging view on Alzheimer disease's (AD) pathogenesis considers amyloid-ß (Aß) oligomers as a key factor in synaptic impairment and rodent spatial memory decline. Alterations in the α7-nicotinic acetylcholine receptor (α7-nAChR) have been implicated in AD pathology. Herein, we report that nicotine, an unselective α7-nAChR agonist, protects from morphological and synaptic impairments induced by Aß oligomers. Interestingly, nicotine prevents both early postsynaptic impairment and late presynaptic damage induced by Aß oligomers through the α7-nAChR/phosphatidylinositol-3-kinase (PI3K) signaling pathway. On the other hand, a cross-talk between α7-nAChR and the Wnt/ß-catenin signaling pathway was revealed by the following facts: (1) nicotine stabilizes ß-catenin, in a concentration-dependent manner; (2) nicotine prevents Aß-induced loss of ß-catenin through the α7-nAChR; and (3) activation of canonical Wnt/ß-catenin signaling induces α7-nAChR expression. Analysis of the α7-nAChR promoter indicates that this receptor is a new Wnt target gene. Taken together, these results demonstrate that nicotine prevents memory deficits and synaptic impairment induced by Aß oligomers. In addition, nicotine improves memory in young APP/PS1 transgenic mice before extensive amyloid deposition and senile plaque development, and also in old mice where senile plaques have already formed. Activation of the α7-nAChR/PI3K signaling pathway and its cross-talk with the Wnt signaling pathway might well be therapeutic targets for potential AD treatments.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/toxicity , Nicotine/pharmacology , Peptide Fragments/toxicity , alpha7 Nicotinic Acetylcholine Receptor/physiology , Amyloid beta-Peptides/chemical synthesis , Amyloid beta-Protein Precursor/genetics , Androstadienes/pharmacology , Animals , Bungarotoxins/pharmacology , Cells, Cultured , Dendrites/drug effects , Dendrites/ultrastructure , Disks Large Homolog 4 Protein , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Intracellular Signaling Peptides and Proteins/analysis , Maze Learning/drug effects , Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurites/ultrastructure , Neurons/drug effects , Neurons/metabolism , Neurons/ultrastructure , Nicotine/therapeutic use , Patch-Clamp Techniques , Peptide Fragments/chemical synthesis , Phosphatidylinositol 3-Kinases/physiology , Plaque, Amyloid/metabolism , Presenilin-1/genetics , Presynaptic Terminals/drug effects , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Signal Transduction , Synapsins/analysis , Wnt Proteins/physiology , Wnt Signaling Pathway , Wortmannin , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/biosynthesis , alpha7 Nicotinic Acetylcholine Receptor/genetics , beta Catenin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...