Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 182
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 533(7603): 407-10, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27120163

ABSTRACT

In salamanders, grafting of a left limb blastema onto a right limb stump yields regeneration of three limbs, the normal limb and two 'supernumerary' limbs. This experiment and other research have shown that the juxtaposition of anterior and posterior limb tissue plus innervation are necessary and sufficient to induce complete limb regeneration in salamanders. However, the cellular and molecular basis of the requirement for anterior-posterior tissue interactions were unknown. Here we have clarified the molecular basis of the requirement for both anterior and posterior tissue during limb regeneration and supernumerary limb formation in axolotls (Ambystoma mexicanum). We show that the two tissues provide complementary cross-inductive signals that are required for limb outgrowth. A blastema composed solely of anterior tissue normally regresses rather than forming a limb, but activation of hedgehog (HH) signalling was sufficient to drive regeneration of an anterior blastema to completion owing to its ability to maintain fibroblast growth factor (FGF) expression, the key signalling activity responsible for blastema outgrowth. In blastemas composed solely of posterior tissue, HH signalling was not sufficient to drive regeneration; however, ectopic expression of FGF8 together with endogenous HH signalling was sufficient. In axolotls, FGF8 is expressed only in the anterior mesenchyme and maintenance of its expression depends on sonic hedgehog (SHH) signalling from posterior tissue. Together, our findings identify key anteriorly and posteriorly localized signals that promote limb regeneration and show that these single factors are sufficient to drive non-regenerating blastemas to complete regeneration with full elaboration of skeletal elements.


Subject(s)
Ambystoma/physiology , Choristoma/metabolism , Extremities/physiology , Fibroblast Growth Factor 8/metabolism , Hedgehog Proteins/metabolism , Regeneration/physiology , Signal Transduction , Animals , Body Patterning/physiology , Fibroblast Growth Factor 8/genetics , Mesoderm/metabolism
2.
Int J Mol Sci ; 21(5)2020 Mar 04.
Article in English | MEDLINE | ID: mdl-32143329

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia worldwide, being characterized by the deposition of senile plaques, neurofibrillary tangles (enriched in the amyloid beta (Aß) peptide and hyperphosphorylated tau (p-tau), respectively) and memory loss. Aging, type 2 diabetes (T2D) and female sex (especially after menopause) are risk factors for AD, but their crosslinking mechanisms remain unclear. Most clinical trials targeting AD neuropathology failed and it remains incurable. However, evidence suggests that effective anti-T2D drugs, such as the GLP-1 mimetic and neuroprotector liraglutide, can be also efficient against AD. Thus, we aimed to study the benefits of a peripheral liraglutide treatment in AD female mice. We used blood and brain cortical lysates from 10-month-old 3xTg-AD female mice, treated for 28 days with liraglutide (0.2 mg/kg, once/day) to evaluate parameters affected in AD (e.g., Aß and p-tau, motor and cognitive function, glucose metabolism, inflammation and oxidative/nitrosative stress). Despite the limited signs of cognitive changes in mature female mice, liraglutide only reduced their cortical Aß1-42 levels. Liraglutide partially attenuated brain estradiol and GLP-1 and activated PKA levels, oxidative/nitrosative stress and inflammation in these AD female mice. Our results support the earlier use of liraglutide as a potential preventive/therapeutic agent against the accumulation of the first neuropathological features of AD in females.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Hypoglycemic Agents/pharmacology , Inflammation/metabolism , Liraglutide/pharmacology , Peptide Fragments/metabolism , Animals , Behavior, Animal , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetes Mellitus, Type 2/metabolism , Estradiol/metabolism , Female , Glucagon-Like Peptide 1/metabolism , Glycolysis , Maze Learning , Memory Disorders , Mice , Neurofibrillary Tangles/metabolism , Nitrosative Stress , Oxidative Stress , Phenotype , Plaque, Amyloid/metabolism
3.
Dev Biol ; 433(2): 262-275, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29198566

ABSTRACT

Axolotls can regenerate complex structures through recruitment and remodeling of cells within mature tissues. Accessing the underlying mechanisms at a molecular resolution is crucial to understand how injury triggers regeneration and how it proceeds. However, gene transformation in adult tissues can be challenging. Here we characterize the use of pseudotyped baculovirus (BV) as an effective gene transfer method both for cells within mature limb tissue and within the blastema. These cells remain competent to participate in regeneration after transduction. We further characterize the effectiveness of BV for gene overexpression studies by overexpressing Shh in the blastema, which yields a high penetrance of classic polydactyly phenotypes. Overall, our work establishes BV as a powerful tool to access gene function in axolotl limb regeneration.


Subject(s)
Ambystoma mexicanum/physiology , Forelimb/physiology , Gene Expression Regulation , Genetic Vectors/genetics , Nucleopolyhedroviruses/genetics , Regeneration/physiology , Transduction, Genetic , Ambystoma mexicanum/genetics , Amputation, Surgical , Animals , Gene Expression Profiling , Genes, Reporter , Genes, Synthetic , Hedgehog Proteins/genetics , Hedgehog Proteins/physiology , Homeodomain Proteins/physiology , Humans , Membrane Glycoproteins/physiology , Mesoderm/cytology , Recombinant Proteins/metabolism , Regeneration/genetics , Transgenes , Viral Envelope Proteins/physiology , Wound Healing/genetics , Wound Healing/physiology
4.
Proc Natl Acad Sci U S A ; 113(5): E654-61, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26787876

ABSTRACT

Group living animals must be able to express different behavior profiles depending on their social status. Therefore, the same genotype may translate into different behavioral phenotypes through socially driven differential gene expression. However, how social information is translated into a neurogenomic response and what are the specific cues in a social interaction that signal a change in social status are questions that have remained unanswered. Here, we show for the first time, to our knowledge, that the switch between status-specific neurogenomic states relies on the assessment of fight outcome rather than just on self- or opponent-only assessment of fighting ability. For this purpose, we manipulated the perception of fight outcome in male zebrafish and measured its impact on the brain transcriptome using a zebrafish whole genome gene chip. Males fought either a real opponent, and a winner and a loser were identified, or their own image on a mirror, in which case, despite expressing aggressive behavior, males did not experience either a victory or a defeat. Massive changes in the brain transcriptome were observed in real opponent fighters, with losers displaying both a higher number of differentially expressed genes and of coexpressed gene modules than winners. In contrast, mirror fighters expressed a neurogenomic state similar to that of noninteracting fish. The genes that responded to fight outcome included immediate early genes and genes involved in neuroplasticity and epigenetic modifications. These results indicate that, even in cognitively simple organisms such as zebrafish, neurogenomic responses underlying changes in social status rely on mutual assessment of fighting ability.


Subject(s)
Brain/metabolism , Transcription, Genetic , Zebrafish/physiology , Animals , Chromosomes , Gene Expression , Promoter Regions, Genetic , Zebrafish/genetics
5.
Biochim Biophys Acta ; 1852(8): 1665-75, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25960150

ABSTRACT

We aimed to investigate mitochondrial function, biogenesis and autophagy in the brain of type 2 diabetes (T2D) and Alzheimer's disease (AD) mice. Isolated brain mitochondria and homogenates from cerebral cortex and hippocampus of wild-type (WT), triple transgenic AD (3xTg-AD) and T2D mice were used to evaluate mitochondrial functional parameters and protein levels of mitochondrial biogenesis, autophagy and synaptic integrity markers, respectively. A significant decrease in mitochondrial respiration, membrane potential and energy levels was observed in T2D and 3xTg-AD mice. Also, a significant decrease in the levels of autophagy-related protein 7 (ATG7) and glycosylated lysosomal membrane protein 1 (LAMP1) was observed in cerebral cortex and hippocampus of T2D and 3xTg-AD mice. Moreover, both brain regions of 3xTg-AD mice present lower levels of nuclear respiratory factor (NRF) 1 while the levels of NRF2 are lower in both brain regions of T2D and 3xTg-AD mice. A decrease in mitochondrial encoded, nicotinamide adenine dinucleotide dehydrogenase subunit 1 (ND1) was also observed in T2D and 3xTg-AD mice although only statistically significant in T2D cortex. Furthermore, a decrease in the levels of postsynaptic density protein 95 (PSD95) in the cerebral cortex of 3xTg-AD mice and in hippocampus of T2D and 3xTg-AD mice and a decrease in the levels of synaptosomal-associated protein 25 (SNAP 25) in the hippocampus of T2D and 3xTg-AD mice were observed suggesting synaptic integrity loss. These results support the idea that alterations in mitochondrial function, biogenesis and autophagy cause synaptic damage in AD and T2D.


Subject(s)
Alzheimer Disease , Autophagy/physiology , Biomarkers/metabolism , Brain , Diabetes Mellitus, Type 2 , Mitochondria/pathology , Synapses/metabolism , Alzheimer Disease/complications , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Male , Mice , Mice, Transgenic , Mitochondria/metabolism
6.
Biochim Biophys Acta ; 1852(7): 1428-41, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25857617

ABSTRACT

Oxidative stress and endoplasmic reticulum (ER) stress have been associated with Alzheimer's disease (AD) progression. In this study we analyzed whether oxidative stress involving changes in Nrf2 and ER stress may constitute early events in AD pathogenesis by using human peripheral blood cells and an AD transgenic mouse model at different disease stages. Increased oxidative stress and increased phosphorylated Nrf2 (p(Ser40)Nrf2) were observed in human peripheral blood mononuclear cells (PBMCs) isolated from individuals with mild cognitive impairment (MCI). Moreover, we observed impaired ER Ca2+ homeostasis and increased ER stress markers in PBMCs from MCI individuals and mild AD patients. Evidence of early oxidative stress defense mechanisms in AD was substantiated by increased p(Ser40)Nrf2 in 3month-old 3xTg-AD male mice PBMCs, and also with increased nuclear Nrf2 levels in brain cortex. However, SOD1 protein levels were decreased in human MCI PBMCs and in 3xTg-AD mice brain cortex; the latter further correlated with reduced SOD1 mRNA levels. Increased ER stress was also detected in the brain cortex of young female and old male 3xTg-AD mice. We demonstrate oxidative stress and early Nrf2 activation in AD human and mouse models, which fails to regulate some of its targets, leading to repressed expression of antioxidant defenses (e.g., SOD-1), and extending to ER stress. Results suggest markers of prodromal AD linked to oxidative stress associated with Nrf2 activation and ER stress that may be followed in human peripheral blood mononuclear cells.


Subject(s)
Alzheimer Disease/metabolism , Endoplasmic Reticulum Stress , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Aged , Aged, 80 and over , Animals , Cells, Cultured , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Cognitive Dysfunction/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
7.
Biochim Biophys Acta ; 1843(6): 1150-61, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24589617

ABSTRACT

Abnormal accumulation of amyloid-ß (Aß) peptide in the brain is a pathological hallmark of Alzheimer's disease (AD). In addition to neurotoxic effects, Aß also damages brain endothelial cells (ECs) and may thus contribute to the degeneration of cerebral vasculature, which has been proposed as an early pathogenic event in the course of AD and is able to trigger and/or potentiate the neurodegenerative process and cognitive decline. However, the mechanisms underlying Aß-induced endothelial dysfunction are not completely understood. Here we hypothesized that Aß impairs protein quality control mechanisms both in the secretory pathway and in the cytosol in brain ECs, leading cells to death. In rat brain RBE4 cells, we demonstrated that Aß1-40 induces the failure of the ER stress-adaptive unfolded protein response (UPR), deregulates the ubiquitin-proteasome system (UPS) decreasing overall proteasome activity with accumulation of ubiquitinated proteins and impairs the autophagic protein degradation pathway due to failure in the autophagic flux, which culminates in cell demise. In conclusion, Aß deregulates proteostasis in brain ECs and, as a consequence, these cells die by apoptosis.


Subject(s)
Amyloid beta-Peptides/pharmacology , Apoptosis , Autophagy , Brain/drug effects , Endothelium, Vascular/drug effects , Proteasome Endopeptidase Complex/metabolism , Animals , Blotting, Western , Brain/metabolism , Brain/pathology , Caspases/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Microtubule-Associated Proteins/metabolism , Rats , Ubiquitin/metabolism , Unfolded Protein Response/drug effects
8.
Biochim Biophys Acta ; 1832(12): 2191-203, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23994613

ABSTRACT

Neurovascular dysfunction arising from endothelial cell damage is an early pathogenic event that contributes to the neurodegenerative process occurring in Alzheimer's disease (AD). Since the mechanisms underlying endothelial dysfunction are not fully elucidated, this study was aimed to explore the hypothesis that brain endothelial cell death is induced upon the sustained activation of the endoplasmic reticulum (ER) stress response by amyloid-beta (Aß) peptide, which deposits in the cerebral vessels in many AD patients and transgenic mice. Incubation of rat brain endothelial cells (RBE4 cell line) with Aß1-40 increased the levels of several markers of ER stress-induced unfolded protein response (UPR), in a time-dependent manner, and affected the Ca(2+) homeostasis due to the release of Ca(2+) from this intracellular store. Finally, Aß1-40 was shown to activate both mitochondria-dependent and -independent apoptotic cell death pathways. Enhanced release of cytochrome c from mitochondria and activation of the downstream caspase-9 were observed in cells treated with Aß1-40 concomitantly with caspase-12 activation. Furthermore, Aß1-40 activated the apoptosis effectors' caspase-3 and promoted the translocation of apoptosis-inducing factor (AIF) to the nucleus demonstrating the involvement of caspase-dependent and -independent mechanisms during Aß-induced endothelial cell death. In conclusion, our data demonstrate that ER stress plays a significant role in Aß1-40-induced apoptotic cell death in brain endothelial cells suggesting that ER stress-targeted therapeutic strategies might be useful in AD to counteract vascular defects and ultimately neurodegeneration.


Subject(s)
Amyloid beta-Peptides/pharmacology , Brain/pathology , Endoplasmic Reticulum Stress/drug effects , Endothelium, Vascular/pathology , Peptide Fragments/pharmacology , Activating Transcription Factor 6/metabolism , Animals , Apoptosis/drug effects , Brain/drug effects , Brain/metabolism , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cytosol/metabolism , Endoplasmic Reticulum Chaperone BiP , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , In Situ Nick-End Labeling , L-Lactate Dehydrogenase/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Necrosis , Poly(ADP-ribose) Polymerases/metabolism , Rats
9.
Hum Mol Genet ; 21(21): 4680-702, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22843496

ABSTRACT

Abnormal presence of autophagic vacuoles is evident in brains of patients with Parkinson's disease (PD), in contrast to the rare detection of autophagosomes in a normal brain. However, the actual cause and pathological significance of these observations remain unknown. Here, we demonstrate a role for mitochondrial metabolism in the regulation of the autophagy-lysosomal pathway in ex vivo and in vitro models of PD. We show that transferring mitochondria from PD patients into cells previously depleted of mitochondrial DNA is sufficient to reproduce the alterations in the autophagic system observed in PD patient brains. Although the initial steps of this pathway are not compromised, there is an increased accumulation of autophagosomes associated with a defective autophagic activity. We prove that this functional decline was originated from a deficient mobilization of autophagosomes from their site of formation toward lysosomes due to disruption in microtubule-dependent trafficking. This contributed directly to a decreased proteolytic flux of α-synuclein and other autophagic substrates. Our results lend strong support for a direct impact of mitochondria in autophagy as defective autophagic clearance ability secondary to impaired microtubule trafficking is driven by dysfunctional mitochondria. We uncover mitochondria and mitochondria-dependent intracellular traffic as main players in the regulation of autophagy in PD.


Subject(s)
Lysosomes/metabolism , Microtubules/metabolism , Mitochondria/metabolism , Parkinson Disease , Aged , Autophagy/physiology , Brain/metabolism , Brain/physiopathology , Cell Differentiation , Cells, Cultured , DNA, Mitochondrial/genetics , Humans , Lysosomes/pathology , Microtubules/pathology , Middle Aged , Mitochondria/pathology , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Protein Transport , Signal Transduction , Vacuoles/metabolism , Vacuoles/pathology , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism
10.
Mol Cell Neurosci ; 52: 1-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23026561

ABSTRACT

Previously we reported that amyloid-ß (Aß) leads to endoplasmic reticulum (ER) stress in cultured cortical neurons and that ER-mitochondria Ca(2+) transfer is involved in Aß-induced apoptotic neuronal cell death. In cybrid cells which recreate the defect in mitochondrial cytochrome c oxidase (COX) activity observed in platelets from Alzheimer's disease (AD) patients, we have shown that mitochondrial dysfunction affects the ER stress response triggered by Aß. Here, we further investigated the impact of COX inhibition on Aß-induced ER dysfunction using a neuronal model. Primary cultures of cortical neurons were challenged with toxic concentrations of Aß upon chemical inhibition of COX with potassium cyanide (KCN). ER Ca(2+) homeostasis was evaluated under these conditions, together with the levels of ER stress markers, namely the chaperone GRP78 and XBP-1, a mediator of the ER unfolded protein response (UPR). We demonstrated that COX inhibition potentiates the Aß-induced depletion of ER Ca(2+) content. KCN pre-treatment was also shown to enhance the rise of cytosolic Ca(2+) levels triggered by Aß and thapsigargin, a widely used ER stressor. This effect was reverted in the presence of dantrolene, an inhibitor of ER Ca(2+) release through ryanodine receptors. Similarly, the increase in GRP78 and XBP-1 protein levels was shown to be higher in neurons treated with Aß or thapsigargin in the presence of KCN in comparison with levels determined in neurons treated with the neurotoxins alone. Although the decrease in cell survival, the activation of caspase-9- and -3-mediated apoptotic cell death observed in Aß- and thapsigargin-treated neurons were also potentiated by KCN, this effect is less pronounced than that observed in Ca(2+) signalling and UPR. Furthermore, in neurons treated with Aß, the potentiating effect of the COX inhibitor in cell survival and death was not prevented by dantrolene. These results show that inhibition of mitochondrial COX activity potentiates Aß-induced ER dysfunction and, to a less extent, neuronal cell death. Furthermore, data supports that the effect of impaired COX on Aß-induced cell death occurs independently of Ca(2+) release through ER ryanodine receptors. Together, our data demonstrate that mitochondria dysfunction in AD enhances the neuronal susceptibility to toxic insults, namely to Aß-induced ER stress, and strongly suggest that the close communication between ER and mitochondria can be a valuable future therapeutic target in AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Apoptosis/physiology , Cerebral Cortex/metabolism , Electron Transport Complex IV/metabolism , Endoplasmic Reticulum Stress/physiology , Mitochondria/metabolism , Neurons/metabolism , Amyloid beta-Peptides/toxicity , Animals , Blotting, Western , Rats , Rats, Wistar
11.
Biochim Biophys Acta ; 1822(2): 139-49, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22037589

ABSTRACT

Machado-Joseph disease (MJD), also known as Spinocerebellar Ataxia type 3, is an inherited dominant autosomal neurodegenerative disorder. An expansion of Cytosine-Adenine-Guanine (CAG) repeats in the ATXN3 gene is translated as an expanded polyglutamine domain in the disease protein, ataxin-3. Selective neurodegeneration in MJD is evident in several subcortical brain regions including the cerebellum. Mitochondrial dysfunction has been proposed as a mechanism of neurodegeneration in polyglutamine disorders. In this study, we used different cell models and transgenic mice to assess the importance of mitochondria on cytotoxicity observed in MJD. Transiently transfected HEK cell lines with expanded (Q84) ataxin-3 exhibited a higher susceptibility to 3-nitropropionic acid (3-NP), an irreversible inhibitor of mitochondrial complex II. Increased susceptibility to 3-NP was also detected in stably transfected PC6-3 cells that inducibly express expanded (Q108) ataxin-3 in a tetracycline-regulated manner. Moreover, cerebellar granule cells from MJD transgenic mice were more sensitive to 3-NP inhibition than wild-type cerebellar neurons. PC6-3 (Q108) cells differentiated into a neuronal-like phenotype with nerve growth factor (NGF) exhibited a significant decrease in mitochondrial complex II activity. Mitochondria from MJD transgenic mouse model and lymphoblast cell lines derived from MJD patients also showed a trend toward reduced complex II activity. Our results suggest that mitochondrial complex II activity is moderately compromised in MJD, which may designate a common feature in polyglutamine toxicity.


Subject(s)
Machado-Joseph Disease/metabolism , Mitochondria/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Animals , Ataxin-3 , Cell Death/genetics , Cell Death/physiology , Cell Line , Cell Line, Transformed , Cells, Cultured , Cerebellum/metabolism , Disease Models, Animal , HEK293 Cells , Humans , Machado-Joseph Disease/genetics , Machado-Joseph Disease/pathology , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondrial Membranes/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neurons/metabolism , Nitro Compounds/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Peptides/metabolism , Propionates/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Tetracycline/pharmacology
12.
Neurobiol Dis ; 49: 1-12, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22940631

ABSTRACT

Intensive insulin therapy can prevent or slow the progression of long-term diabetes complications but, at the same time, it increases the risk for episodes of severe hypoglycemia. In our study, we used a protocol intended to mimic the levels of blood glucose that occur in type 1 diabetic patients under an intensive insulin therapy. Streptozotocin (STZ)-induced diabetic rats were treated subcutaneously with twice-daily insulin injections for 2weeks to induce hypoglycemic episodes. Brain cortical and hippocampal mitochondria were isolated and mitochondrial bioenergetics (respiratory chain and phosphorylation system) and oxidative status parameters (malondialdehyde (MDA) levels, mitochondrial aconitase activity and enzymatic and non-enzymatic antioxidant defenses) were analyzed. The protein levels of synaptophysin, a marker of synaptic integrity, and caspase 9 activity were also evaluated in cortical and hippocampal homogenates. Brain cortical mitochondria isolated from hyper- and recurrent hypoglycemic animals presented higher levels of MDA and α-tocopherol together with an increased glutathione disulfide reductase activity, lower manganese superoxide dismutase (MnSOD) activity and glutathione-to-glutathione disulfide (GSH/GSSG) ratio. No significant alterations were found in cortical mitochondrial respiratory chain and oxidative phosphorylation system. Hippocampal mitochondria from both experimental groups presented an impaired oxidative phosphorylation system characterized by a decreased mitochondrial energization potential and ATP levels and higher repolarization lag phase. In addition, higher MDA levels and decreased GSH/GSSG, α-tocopherol levels, and aconitase, glutathione peroxidase and MnSOD activities were observed in both groups of animals. Hippocampal mitochondria from recurrent hypoglycemic animals also showed an impairment of the respiratory chain characterized by a lower state 3 of respiration, respiratory control ratio and ADP/O index, and a higher state 4 of respiration. Additionally, a non-statistically significant decrease in synaptophysin protein levels was observed in cortical homogenates from recurrent hypoglycemic rats as well as in hippocampal homogenates from hyperglycemic and recurrent hypoglycemic rats. An increase in caspase 9 activity was also observed in hippocampal homogenates from hyperglycemic and recurrent hypoglycemic animals. Our results show that mitochondrial dysfunction induced by long-term hyperglycemic effects is exacerbated by recurrent hypoglycemia, which may compromise the function and integrity of brain cells.


Subject(s)
Brain/metabolism , Diabetes Mellitus, Experimental/metabolism , Hypoglycemia/metabolism , Hypoglycemic Agents/adverse effects , Insulin/adverse effects , Mitochondria/metabolism , Aconitate Hydratase/metabolism , Animals , Brain/drug effects , Caspase 9/metabolism , Energy Metabolism/drug effects , Energy Metabolism/physiology , Glutathione/metabolism , Glutathione Disulfide/metabolism , Glutathione Peroxidase/metabolism , Hypoglycemia/chemically induced , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Male , Malondialdehyde/metabolism , Mitochondria/drug effects , Random Allocation , Rats, Wistar , Superoxide Dismutase/metabolism , Synaptophysin/metabolism , alpha-Tocopherol/metabolism
13.
Neurochem Res ; 38(4): 797-806, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23389658

ABSTRACT

Alzheimer's disease, the most prevalent age-related neurodegenerative disease, is characterized by the presence of extracellular senile plaques composed of amyloid-beta (Aß) peptide and intracellular neurofibrillary tangles. More than 50 % of Alzheimer's disease (AD) patients also exhibit abundant accumulation of α-synuclein (α-Syn)-positive Lewy bodies. This Lewy body variant of AD (LBV-AD) is associated with accelerated cognitive dysfunction and progresses more rapidly than pure AD. In addition, it has been suggested that Aß and α-Syn can directly interact. In this study we investigated the effect of α-Syn on Aß-induced toxicity in cortical neurons. In order to mimic the intracellular accumulation of α-Syn observed in the brain of LBV-AD patients, we used valproic acid (VPA) to increase its endogenous expression levels. The release of α-Syn from damaged presynaptic terminals that occurs during the course of the disease was simulated by challenging cells with recombinant α-Syn. Our results showed that either VPA-induced α-Syn upregulation or addition of recombinant α-Syn protect primary cortical neurons from soluble Aß1-42 decreasing the caspase-3-mediated cell death. It was also found that neuroprotection against Aß-induced toxicity mediated by α-Syn overexpression involves the PI3K/Akt cell survival pathway. Furthermore, recombinant α-Syn was shown to directly interact with Aß1-42 and to decrease the levels of Aß1-42 oligomers, which might explain its neuroprotective effect. In conclusion, we demonstrate that either endogenous or exogenous α-Syn can be neuroprotective against Aß-induced cell death, suggesting a cell defence mechanism during the initial stages of the mixed pathology.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/toxicity , Neurons/drug effects , Peptide Fragments/toxicity , alpha-Synuclein/pharmacology , Amyloid beta-Peptides/metabolism , Animals , Caspase 3/metabolism , Cerebral Cortex/cytology , Neurons/metabolism , Peptide Fragments/metabolism , Rats , Valproic Acid/pharmacology , alpha-Synuclein/metabolism
14.
Methods Mol Biol ; 2562: 369-387, 2023.
Article in English | MEDLINE | ID: mdl-36272088

ABSTRACT

Salamanders have served as an excellent model for developmental and tissue regeneration studies. While transgenic approaches are available for various salamander species, their long generation time and expensive maintenance have driven the development of alternative gene delivery methods for functional studies. We have previously developed pseudotyped baculovirus (BV) as a tool for gene delivery in the axolotl (Oliveira et al. Dev Biol 433(2):262-275, 2018). Since its initial conception, we have refined our protocol of BV production and usage in salamander models. In this chapter, we describe a detailed and versatile protocol for BV-mediated transduction in urodeles.


Subject(s)
Ambystoma mexicanum , Baculoviridae , Animals , Ambystoma mexicanum/genetics , Baculoviridae/genetics , Animals, Genetically Modified , Urodela
15.
Dev Cell ; 58(22): 2416-2427.e7, 2023 11 20.
Article in English | MEDLINE | ID: mdl-37879337

ABSTRACT

Axolotl limb regeneration is accompanied by the transient induction of cellular senescence within the blastema, the structure that nucleates regeneration. The precise role of this blastemal senescent cell (bSC) population, however, remains unknown. Here, through a combination of gain- and loss-of-function assays, we elucidate the functions and molecular features of cellular senescence in vivo. We demonstrate that cellular senescence plays a positive role during axolotl regeneration by creating a pro-proliferative niche that supports progenitor cell expansion and blastema outgrowth. Senescent cells impact their microenvironment via Wnt pathway modulation. Further, we identify a link between Wnt signaling and senescence induction and propose that bSC-derived Wnt signals facilitate the proliferation of neighboring cells in part by preventing their induction into senescence. This work defines the roles of cellular senescence in the regeneration of complex structures.


Subject(s)
Ambystoma mexicanum , Cellular Senescence , Animals , Ambystoma mexicanum/metabolism , Wnt Signaling Pathway , Stem Cells , Cell Proliferation , Extremities
16.
Neurobiol Dis ; 45(1): 206-18, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21854848

ABSTRACT

The current study was undertaken to address the role of mitochondrial reactive oxygen species (ROS), and hypoxia inducible factor-1 alpha (HIF-1α) signaling pathway in the protection against high glucose levels in brain endothelial and NT2 neuron-like cells. Rat brain endothelial cells (RBE4) treated with non-toxic concentrations of cyanide (≤1 µM; 1h) exhibited an increase in ROS levels, particularly hydrogen peroxide (H(2)O(2)). Cyanide also induced a modest mitochondrial depolarization, an increase in oxygen consumption and a structural (smaller mitochondria) and spatial (perinuclear region) reorganization of mitochondrial network. The stabilization and nuclear activation of HIF-1α in the presence of cyanide were also observed, which resulted in an increase in vascular endothelial growth factor (VEGF), endothelial nitric oxide synthase (eNOS) and erythropoietin (EPO) protein levels reflecting an adaptive response. Importantly, preconditioning induced by cyanide protected brain endothelial cells against high glucose-mediated damage by the prevention of apoptotic cell death. In mitochondrial DNA-depleted NT2 (NT2 ρ0) cells, cyanide (0.1 µM) was unable to stimulate ROS production and, consequently, protect against glucotoxicity. Conversely, in NT2 cells, the parental cells with functional mitochondria, cyanide significantly increased ROS levels protecting against high glucose-induced neuronal cell loss and activation of caspase-3. The free radical scavenger N-acetyl-L-cysteine and the specific HIF-1α inhibitor 2-methoxyestradiol completely abolished the protective effects of cyanide preconditioning. Altogether our results demonstrate that mitochondrial preconditioning induced by cyanide triggers a protective response mediated by mitochondrial ROS and HIF-1α activation and signaling, which render brain endothelial and neuronal cells resistant against glucotoxicity.


Subject(s)
Brain/drug effects , Endothelial Cells/drug effects , Glucose/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondria/drug effects , Neurons/drug effects , Potassium Cyanide/pharmacology , Reactive Oxygen Species/metabolism , Animals , Brain/cytology , Brain/metabolism , Caspase 3/metabolism , Cell Line , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Hydrogen Peroxide/metabolism , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Neurons/cytology , Neurons/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , Rats , Vascular Endothelial Growth Factor A/metabolism
17.
Nat Commun ; 13(1): 1141, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35241664

ABSTRACT

Salamander limb regeneration is an accurate process which gives rise exclusively to the missing structures, irrespective of the amputation level. This suggests that cells in the stump have an awareness of their spatial location, a property termed positional identity. Little is known about how positional identity is encoded, in salamanders or other biological systems. Through single-cell RNAseq analysis, we identified Tig1/Rarres1 as a potential determinant of proximal identity. Tig1 encodes a conserved cell surface molecule, is regulated by retinoic acid and exhibits a graded expression along the proximo-distal axis of the limb. Its overexpression leads to regeneration defects in the distal elements and elicits proximal displacement of blastema cells, while its neutralisation blocks proximo-distal cell surface interactions. Critically, Tig1 reprogrammes distal cells to a proximal identity, upregulating Prod1 and inhibiting Hoxa13 and distal transcriptional networks. Thus, Tig1 is a central cell surface determinant of proximal identity in the salamander limb.


Subject(s)
Extremities , Urodela , Amputation, Surgical , Animals , Extremities/physiology , Tretinoin/pharmacology , Urodela/genetics
18.
Synapse ; 65(6): 457-66, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20853444

ABSTRACT

In this work, we evaluated the effects of streptozotocin (STZ)-induced hyperglycemia and an acute episode of insulin-induced hypoglycemia in plasma amino acids and cortical neurotransmitters. For that purpose, we used citrate (vehicle)-treated Wistar rats, STZ-treated rats [i.p., 50 mg/kg body weight], and STZ-treated rats injected with insulin [s.c., dose adjusted with blood glucose levels] 1 h prior to sacrifice to induce an acute episode of hypoglycemia. Plasma was collected for determination of amino acids levels. In addition, cortical synaptosomal preparations were obtained and the total levels of neurotransmitters, levels of aspartate, glutamate, taurine, and GABA released by the action of KCl, iodoacetic acid (IAA), ouabain, and veratridine, membrane potential and ATP levels were evaluated. Compared with control rats, plasma from hypoglycemic rats presented increased levels of aspartate, glutamate, glutamine, and taurine whereas GABA levels were decreased in STZ and hypoglycemic rats. Similarly, glutamate and taurine levels were increased in hypoglycemic synaptosomes while GABA decreased in hypoglycemic and STZ-diabetic synaptosomes. The depolarizing agent KCl promoted an increase in aspartate, glutamate, and taurine release from hypoglycemic synaptosomes. The highest release of neurotransmitters occurred in the presence of veratridine and ouabain, two other depolarizing agents, in all groups of experimental animals. However, a higher release of glutamate was observed in the diabetic and hypoglycemic synaptosomes. No alterations were observed in synaptosomal membrane potential and ATP levels. These results show that in the presence of a metabolic insult a higher release of excitatory amino acids occurs, which may underlay the neuronal injury observed in type 1 diabetic patients under insulin therapy.


Subject(s)
Amino Acids/blood , Cerebral Cortex/metabolism , Diabetes Mellitus, Experimental/complications , Hyperglycemia/etiology , Hypoglycemia/etiology , Neurotransmitter Agents/blood , Synaptosomes/metabolism , Synaptosomes/pathology , Animals , Cerebral Cortex/pathology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/pathology , Disease Models, Animal , Hyperglycemia/blood , Hyperglycemia/pathology , Hypoglycemia/blood , Hypoglycemia/pathology , Male , Neurotransmitter Agents/metabolism , Rats , Rats, Wistar , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
19.
Handb Exp Pharmacol ; (203): 331-56, 2011.
Article in English | MEDLINE | ID: mdl-21484578

ABSTRACT

A growing body of evidence suggests that mitochondrial abnormalities are involved in diabetes and associated complications. This chapter gives an overview about the effects of diabetes in mitochondrial function of several tissues including the pancreas, skeletal and cardiac muscle, liver, and brain. The realization that mitochondria are at the intersection of cells' life and death has made them a promising target for drug discovery and therapeutic interventions. Here, we also discuss literature that examined the potential protective effect of insulin, insulin-sensitizing drugs, and mitochondrial-targeted antioxidants.


Subject(s)
Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Hypoglycemic Agents/pharmacology , Mitochondria/metabolism , Animals , Antioxidants/pharmacology , Humans , Insulin/metabolism , Insulin Resistance , Mitochondria/drug effects , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/metabolism , Pancrelipase/metabolism
20.
J Enzyme Inhib Med Chem ; 26(4): 485-97, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21067438

ABSTRACT

Because of the complex cascade of molecular events that can occur in the brain of an Alzheimer's disease (AD) patient, the therapy of this neurodegenerative disease seems more likely to be achieved by multifunctional drugs. Herein, a new series of dual-targeting ligands have been developed and in vitro bioevaluated. Their architecture is based on conjugating the acetylcholinesterase inhibition and anti-oxidant properties in one molecular entity. Specifically, a series of naturally occurring phenolic acids with recognized anti-oxidant properties (derivatives of caffeic acid, rosmarinic acid, and trolox) have been conjugated with choline to account for the recognition by acetylcholinesterase (AChE). The synthesized hybrid compounds evidenced AChE inhibitory capacity of micromolar range (rationalized by molecular modeling studies) and good antioxidant properties. Their effects on human neuroblastoma cells, previously treated with beta-amyloid peptides and 1-methyl-4-phenylpyridinium ion neurotoxins (to simulate AD and Parkinson's disease, respectively), also demonstrated a considerable capacity for protection against the cytotoxicity of these stressors.


Subject(s)
Acetylcholinesterase/metabolism , Antioxidants/pharmacology , Choline/pharmacology , Cholinesterase Inhibitors/pharmacology , Phenols/pharmacology , Antioxidants/chemistry , Cell Death/drug effects , Cell Survival/drug effects , Choline/chemistry , Cholinesterase Inhibitors/chemistry , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Humans , Models, Molecular , Molecular Structure , Phenols/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL