Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Cell Mol Life Sci ; 79(9): 487, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35984507

ABSTRACT

Diabetes has been associated with an increased risk of cognitive decline and dementia. However, the mechanisms underlying this association remain unclear and no effective therapeutic interventions exist. Accumulating evidence demonstrates that mitochondrial defects are a key feature of diabetes contributing to neurodegenerative events. It has also been demonstrated that the putative tumor suppressor WW domain-containing oxidoreductase 1 (WWOX) can interact with mitochondria in several pathological conditions. However, its role in diabetes-associated neurodegeneration remains unknown. So, this study aimed to evaluate the role of WWOX activation in high glucose-induced neuronal damage and death. Our experiments were mainly performed in differentiated SH-SY5Y neuroblastoma cells exposed to high glucose and treated (or not) with Zfra1-31, the specific inhibitor of WWOX. Several parameters were analyzed namely cell viability, WWOX activation (tyrosine 33 residue phosphorylation), mitochondrial function, reactive oxygen species (ROS) production, biogenesis, and dynamics, autophagy and oxidative stress/damage. The levels of the neurotoxic proteins amyloid ß (Aß) and phosphorylated Tau (pTau) and of synaptic integrity markers were also evaluated. We observed that high glucose increased the levels of activated WWOX. Interestingly, brain cortical and hippocampal homogenates from young (6-month old) diabetic GK rats showed increased levels of activated WWOX compared to older GK rats (12-month old) suggesting that WWOX plays an early role in the diabetic brain. In neuronal cells, high glucose impaired mitochondrial respiration, dynamics and biogenesis, increased mitochondrial ROS production and decreased mitochondrial membrane potential and ATP production. More, high glucose augmented oxidative stress/damage and the levels of Aß and pTau proteins and affected autophagy, contributing to the loss of synaptic integrity and cell death. Of note, the activation of WWOX preceded mitochondrial dysfunction and cell death. Importantly, the inhibition of WWOX with Zfra1-31 reversed, totally or partially, the alterations promoted by high glucose. Altogether our observations demonstrate that under high glucose conditions WWOX activation contributes to mitochondrial anomalies and neuronal damage and death, which suggests that WWOX is a potential therapeutic target for early interventions. Our findings also support the efficacy of Zfra1-31 in treating hyperglycemia/diabetes-associated neurodegeneration.


Subject(s)
Amyloid beta-Peptides , Mitochondria , Neuroblastoma , WW Domain-Containing Oxidoreductase , Animals , Humans , Rats , Amyloid beta-Peptides/metabolism , Glucose/metabolism , Glucose/pharmacology , Homeostasis , Mitochondria/metabolism , Neuroblastoma/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , WW Domain-Containing Oxidoreductase/genetics , WW Domain-Containing Oxidoreductase/metabolism
2.
Cell Mol Neurobiol ; 42(1): 3-21, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34510330

ABSTRACT

Oxygen sensing and homeostasis is indispensable for the maintenance of brain structural and functional integrity. Under low-oxygen tension, the non-diseased brain has the ability to cope with hypoxia by triggering a homeostatic response governed by the highly conserved hypoxia-inducible family (HIF) of transcription factors. With the advent of advanced neuroimaging tools, it is now recognized that cerebral hypoperfusion, and consequently hypoxia, is a consistent feature along the Alzheimer's disease (AD) continuum. Of note, the reduction in cerebral blood flow and tissue oxygenation detected during the prodromal phases of AD, drastically aggravates as disease progresses. Within this scenario a fundamental question arises: How HIF-driven homeostatic brain response to hypoxia "behaves" during the AD continuum? In this sense, the present review is aimed to critically discuss and summarize the current knowledge regarding the involvement of hypoxia and HIF signaling in the onset and progression of AD pathology. Importantly, the promises and challenges of non-pharmacological and pharmacological strategies aimed to target hypoxia will be discussed as a new "hope" to prevent and/or postpone the neurodegenerative events that occur in the AD brain.


Subject(s)
Alzheimer Disease , Alzheimer Disease/pathology , Brain/pathology , Cerebrovascular Circulation , Humans , Neuroimaging , Oxygen
3.
Int J Mol Sci ; 22(1)2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33466445

ABSTRACT

The lack of effective disease-modifying therapeutics to tackle Alzheimer's disease (AD) is unsettling considering the actual prevalence of this devastating neurodegenerative disorder worldwide. Intermittent hypoxic conditioning (IHC) is a powerful non-pharmacological procedure known to enhance brain resilience. In this context, the aim of the present study was to investigate the potential long-term protective impact of IHC against AD-related phenotype, putting a special focus on cognition and mitochondrial bioenergetics and dynamics. For this purpose, six-month-old male triple transgenic AD mice (3×Tg-AD) were submitted to an IHC protocol for two weeks and the behavioral assessment was performed at 8.5 months of age, while the sacrifice of mice occurred at nine months of age and their brains were removed for the remaining analyses. Interestingly, IHC was able to prevent anxiety-like behavior and memory and learning deficits and significantly reduced brain cortical levels of amyloid-ß (Aß) in 3×Tg-AD mice. Concerning brain energy metabolism, IHC caused a significant increase in brain cortical levels of glucose and a robust improvement of the mitochondrial bioenergetic profile in 3×Tg-AD mice, as mirrored by the significant increase in mitochondrial membrane potential (ΔΨm) and respiratory control ratio (RCR). Notably, the improvement of mitochondrial bioenergetics seems to result from an adaptative coordination of the distinct but intertwined aspects of the mitochondrial quality control axis. Particularly, our results indicate that IHC favors mitochondrial fusion and promotes mitochondrial biogenesis and transport and mitophagy in the brain cortex of 3×Tg-AD mice. Lastly, IHC also induced a marked reduction in synaptosomal-associated protein 25 kDa (SNAP-25) levels and a significant increase in both glutamate and GABA levels in the brain cortex of 3×Tg-AD mice, suggesting a remodeling of the synaptic microenvironment. Overall, these results demonstrate the effectiveness of the IHC paradigm in forestalling the AD-related phenotype in the 3×Tg-AD mouse model, offering new insights to AD therapy and forcing a rethink concerning the potential value of non-pharmacological interventions in clinical practice.


Subject(s)
Alzheimer Disease/physiopathology , Cognition Disorders/physiopathology , Cognition/physiology , Energy Metabolism/physiology , Hypoxia/physiopathology , Mice, Transgenic/physiology , Mitochondria/physiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Anxiety/metabolism , Anxiety/physiopathology , Brain/metabolism , Brain/physiopathology , Cognition Disorders/metabolism , Disease Models, Animal , Hypoxia/metabolism , Male , Mice , Mice, Transgenic/metabolism , Mitochondria/metabolism
4.
Adv Exp Med Biol ; 1128: 161-183, 2019.
Article in English | MEDLINE | ID: mdl-31062330

ABSTRACT

Due to the exponential growth of aging population worldwide, neurodegenerative diseases became a major public health concern. Among them, Alzheimer's disease (AD) prevails as the most common in the elderly, rendering it a research priority. After several decades considering the brain as an insulin-insensitive organ, recent advances proved a central role for this hormone in learning and memory processes and showed that AD shares a high number of features with systemic conditions characterized by insulin resistance. Mitochondrial dysfunction has also been widely demonstrated to play a major role in AD development supporting the idea that this neurodegenerative disease is characterized by a pronounced metabolic dysregulation. This chapter is intended to discuss evidence demonstrating the key role of insulin signaling and mitochondrial anomalies in AD.


Subject(s)
Alzheimer Disease/pathology , Insulin Resistance , Insulin/physiology , Mitochondria/pathology , Signal Transduction , Humans
5.
Biochim Biophys Acta ; 1862(10): 1909-17, 2016 10.
Article in English | MEDLINE | ID: mdl-27460705

ABSTRACT

The vigorous axonal transport of mitochondria, which serves to distribute these organelles in a dynamic and non-uniform fashion, is crucial to fulfill neuronal energetic requirements allowing the maintenance of neurons structure and function. Particularly, axonal transport of mitochondria and their spatial distribution among the synapses are directly correlated with synaptic activity and integrity. Despite the basis of Alzheimer's disease (AD) remains enigmatic, axonal pathology and synaptic dysfunction occur prior the occurrence of amyloid-ß (Aß) deposition and tau aggregation, the two classical hallmarks of this devastating neurodegenerative disease. Importantly, the early stages of AD are marked by defects on axonal transport of mitochondria as denoted by the abnormal accumulation of mitochondria within large swellings along dystrophic and degenerating neuritis. Within this scenario, this review is devoted to identify the molecular "roadblocks" underlying the abnormal axonal transport of mitochondria and consequent synaptic "starvation" and neuronal degeneration in AD. Understanding the molecular nature of defective mitochondrial transport may provide a new avenue to counteract AD pathology.


Subject(s)
Alzheimer Disease/metabolism , Axons/metabolism , Mitochondria/metabolism , Neuritis/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Axons/pathology , Biological Transport, Active , Humans , Mitochondria/pathology , Neuritis/pathology
6.
Handb Exp Pharmacol ; 240: 281-308, 2017.
Article in English | MEDLINE | ID: mdl-28251365

ABSTRACT

Alzheimer's disease (AD) is a difficult puzzle to solve, in part because the etiology of this devastating neurodegenerative disorder remains murky. However, diabetes has been pinpointed as a major risk factor for the sporadic forms of AD. Several overlapping neurodegenerative mechanisms have been identified between AD and diabetes, including mitochondrial malfunction. This is not surprising taking into account that neurons are cells with a complex morphology, long lifespan, and high energetic requirements which make them particularly reliant on a properly organized and dynamic mitochondrial network to sustain neuronal function and integrity. In this sense, this chapter provides an overview on the role of mitochondrial bioenergetics and dynamics to the neurodegenerative events that occur in AD and diabetes, and how these organelles may represent a mechanistic link between these two pathologies. From a therapeutic perspective, it will be discussed how mitochondria can be targeted in order to efficaciously counteract neurodegeneration associated with AD and diabetes.


Subject(s)
Alzheimer Disease/etiology , Diabetic Neuropathies/etiology , Mitochondria/physiology , Neurodegenerative Diseases/etiology , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Animals , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/physiopathology , Energy Metabolism , Humans , Mitochondria/drug effects , Mitochondrial Dynamics , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/physiopathology
7.
J Neural Transm (Vienna) ; 123(2): 107-11, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25608860

ABSTRACT

Multiple lines of evidence suggest that vascular alterations contribute to Alzheimer's disease (AD) pathogenesis. It is also well established that mitochondrial abnormalities occur early in course of AD. Here, we give an overview of the vascular and mitochondrial abnormalities occurring in AD, including mitochondrial alterations in vascular endothelial cells within the brain, which is emerging as a common feature that bridges cerebral vasculature and mitochondrial metabolism.


Subject(s)
Alzheimer Disease/physiopathology , Brain/physiopathology , Cerebrovascular Circulation/physiology , Mitochondria/physiology , Alzheimer Disease/pathology , Animals , Brain/pathology , Humans , Mitochondria/pathology
8.
J Bioenerg Biomembr ; 47(1-2): 119-31, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25217852

ABSTRACT

The integrity of mitochondrial function is essential to cell life. It follows that disturbances of mitochondrial function will lead to disruption of cell function, expressed as disease or even death. Considering that neuronal uncoupling proteins (UCPs) decrease reactive oxygen species (ROS) production at the expense of energy production, it is important to understand the underlying mechanisms by which UCPs control the balance between the production of adenosine triphosphate (ATP) and ROS in the context of normal physiological activity and in pathological conditions. Here we review the current understanding of neuronal UCPs-mediated respiratory uncoupling process by performing a survey in their physiology and regulation. The latest findings regarding neuronal UCPs physiological roles and their involvement and interest as potential targets for therapeutic intervention in brain diseases will also be exploited.


Subject(s)
Brain Diseases/metabolism , Brain/metabolism , Ion Channels/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Neurons/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain/pathology , Brain Diseases/drug therapy , Brain Diseases/pathology , Energy Metabolism , Humans , Mitochondria/pathology , Neurons/pathology , Reactive Oxygen Species/metabolism , Uncoupling Protein 1
9.
Biomedicines ; 12(3)2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38540092

ABSTRACT

Alzheimer's disease (AD) is a progressive and multifactorial disease that significantly compromises the lives of millions of people worldwide [...].

10.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167222, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38729530

ABSTRACT

Alzheimer's disease (AD) research started several decades ago and despite the many efforts employed to develop new treatments or approaches to slow and/or revert disease progression, AD treatment remains an unsolved issue. Knowing that mitochondria loss of function is a central hub for many AD-associated pathophysiological processes, there has been renewed interest in exploring mitochondria as targets for intervention. In this perspective, the present study was aimed to investigate the possible beneficial effects of 2,4 dinitrophenol (DNP), a mitochondrial uncoupler agent, in an in vitro model of AD. Retinoic acid-induced differentiated SH-SY5Y cells were incubated with okadaic acid (OA), a neurotoxin often used as an AD experimental model, and/or with DNP. OA caused a decrease in neuronal cells viability, induced multiple mitochondrial anomalies including increased levels of reactive oxygen species, decreased bioenergetics and mitochondria content markers, and an altered mitochondria morphology. OA-treated cells also presented increased lipid peroxidation levels, and overactivation of tau related kinases (GSK3ß, ERK1/2 and AMPK) alongside with a significant augment in tau protein phosphorylation levels. Interestingly, DNP co-treatment ameliorated and rescued OA-induced detrimental effects not only on mitochondria but also but also reinstated signaling pathways homeostasis and ameliorated tau pathology. Overall, our results show for the first time that DNP has the potential to preserve mitochondria homeostasis under a toxic insult, like OA exposure, as well as to reestablish cellular signaling homeostasis. These observations foster the idea that DNP, as a mitochondrial modulator, might represent a new avenue for treatment of AD.


Subject(s)
2,4-Dinitrophenol , Alzheimer Disease , Mitochondria , Okadaic Acid , Reactive Oxygen Species , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Okadaic Acid/pharmacology , Okadaic Acid/toxicity , Humans , 2,4-Dinitrophenol/pharmacology , Mitochondria/metabolism , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Cell Survival/drug effects , tau Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Lipid Peroxidation/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Tretinoin/pharmacology
11.
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
12.
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
13.
IUBMB Life ; 64(12): 951-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23129399

ABSTRACT

Alterations of the insulin signaling cascade underlie cognitive decline and the development of several neurodegenerative diseases. In recent years, a great interest has been put in studying the interaction between diabetes and Alzheimer's disease (AD). In fact, evidence shows that both diseases present several biochemical similarities including defects in the insulin signaling pathway. Here, we give an overview of the main functions of insulin in the central nervous system. The impact of insulin signaling impairment in brain aging and AD is also discussed. Finally, we present evidence supporting the notion that insulin is a link between diabetes and AD.


Subject(s)
Aging , Alzheimer Disease/metabolism , Brain/metabolism , Diabetes Mellitus/metabolism , Insulin/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Brain/pathology , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Gene Expression , Glucose/metabolism , Humans , Insulin/genetics , Insulin Resistance , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Neurons/metabolism , Neurons/pathology , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Risk Factors , Signal Transduction
14.
Adv Exp Med Biol ; 724: 205-21, 2012.
Article in English | MEDLINE | ID: mdl-22411245

ABSTRACT

Mitochondria have been long known as "gatekeepers of life and death". Indeed, these dynamic organelles are the master coordinators of energy metabolism, being responsible for the generation of the majority of cellular ATP. Notably, mitochondria are also one of the primary producers of intracellular reactive oxygen species which are the main inducer of oxidative damage. Neurons, as metabolically active cells with high energy demands, are predominantly dependent on mitochondrial function, as reflected by the observation that mitochondrial defects are key features of chronic neurodegenerative diseases. Indeed, morphologic, biochemical and molecular genetic studies posit that mitochondria constitute a convergence point for neurodegeneration. Moreover, recent findings convey that neurons are particularly reliant on the dynamic properties of mitochondria, further emphasizing the critical role of mitochondria in neuronal functions. This chapter highlights how mitochondrial pathobiology might contribute to neurodegeneration in Alzheimer's, Parkinson's and Huntington's diseases.


Subject(s)
Huntington Disease/etiology , Mitochondrial Diseases/complications , Neurodegenerative Diseases/etiology , Parkinson Disease/etiology , Humans , Huntington Disease/pathology , Mitochondria/metabolism , Mitochondria/pathology , Neurodegenerative Diseases/pathology , Parkinson Disease/pathology
15.
Antioxid Redox Signal ; 37(10-12): 739-757, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35316086

ABSTRACT

Aims: Brief episodes of sublethal hypoxia reprogram brain response to face possible subsequent lethal stimuli by triggering adaptive and prosurvival events-a phenomenon denominated hypoxic preconditioning (HP). To date, the potential therapeutic implications of HP to forestall sporadic Alzheimer's disease (sAD) pathology remain unexplored. Using a well-established protocol of HP and focusing on hippocampus as a first brain region affected in AD, this study was undertaken to investigate the potential protective effects of HP in a sAD rat model induced by the intracerebroventricular (icv) administration of streptozotocin (STZ) and to uncover the mitochondrial adaptations underlying this nonpharmacological strategy. Results: HP prevented the memory and learning deficits as well as tau pathology in the icvSTZ rat model. HP also attenuated icvSTZ-related reactive astrogliosis, as noted by increased glial fibrillary acidic protein immunoreactivity and myo-inositol levels. Notably, HP abrogated the icvSTZ-related impaired energy metabolism and oxidative damage. Particularly, HP averted increased lactate, glutamate, and succinate levels, and decreased mitochondrial respiratory chain function and mitochondrial DNA content. Concerning mitochondrial adaptations underlying HP-triggered tolerance to icvSTZ, preconditioned hippocampal mitochondria displayed an enhanced complex II-energized mitochondrial respiration, which resulted from a coordinated interaction between mitochondrial biogenesis and fusion-fission. Mitochondrial biogenesis was stimulated immediately after HP, whereas in a latter phase mitochondrial fusion-fission events are modulated favoring the generation of elongated mitochondria. Innovation and Conclusion: Overall, these results demonstrate for the first time that HP prevents the sAD-like phenotype, in part, by targeting mitochondria emerging as a preventive strategy in the context of AD. Antioxid. Redox Signal. 37, 739-757.


Subject(s)
Alzheimer Disease , Hypoxia , Mitochondria , Alzheimer Disease/metabolism , Alzheimer Disease/prevention & control , Animals , DNA, Mitochondrial/metabolism , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Glutamates/metabolism , Hypoxia/metabolism , Inositol/metabolism , Lactates/metabolism , Mitochondria/metabolism , Phenotype , Rats , Streptozocin , Succinates/metabolism
16.
Commun Biol ; 5(1): 1220, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36357561

ABSTRACT

We conducted a genome-wide association study in a large population of infertile men due to unexplained spermatogenic failure (SPGF). More than seven million genetic variants were analysed in 1,274 SPGF cases and 1,951 unaffected controls from two independent European cohorts. Two genomic regions were associated with the most severe histological pattern of SPGF, defined by Sertoli cell-only (SCO) phenotype, namely the MHC class II gene HLA-DRB1 (rs1136759, P = 1.32E-08, OR = 1.80) and an upstream locus of VRK1 (rs115054029, P = 4.24E-08, OR = 3.14), which encodes a protein kinase involved in the regulation of spermatogenesis. The SCO-associated rs1136759 allele (G) determines a serine in the position 13 of the HLA-DRß1 molecule located in the antigen-binding pocket. Overall, our data support the notion of unexplained SPGF as a complex trait influenced by common variation in the genome, with the SCO phenotype likely representing an immune-mediated condition.


Subject(s)
Genome-Wide Association Study , Infertility, Male , Humans , Male , Infertility, Male/genetics , Spermatogenesis/genetics , Sertoli Cells/metabolism , Alleles , Protein Serine-Threonine Kinases , Intracellular Signaling Peptides and Proteins/metabolism
17.
J Neurochem ; 117(6): 927-36, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21447003

ABSTRACT

Rapamycin is a macrolide immunosuppressant drug, originally used as an anti-fungal agent, which is widely used in transplantation medicine to prevent organ rejection. Target of rapamycin (TOR) is an evolutionarily conserved serine/threonine kinase with pleiotropic cellular functions, regulating processes such as growth and metabolism, cell survival, transcription and autophagy. TOR intervenes in two distinct enzymatic complexes with different functions, a rapamycin-sensitive complex TORC1 and a rapamycin-insensitive complex TORC2. Rapamycin has an inhibitory effect on TORC1 activity and it has been suggested to increase life span, an effect correlated with decreased protein biosynthesis and autophagy activation. In the CNS, rapamycin shows beneficial effects in neuronal survival and plasticity, thus contributing to memory improvement. In this review, evidence implying rapamycin and TOR in aging/life span extension and memory improvement will be discussed. Recent findings about the effects of rapamycin on Alzheimer's disease-associated neuropathology will be also discussed.


Subject(s)
Aging/physiology , Alzheimer Disease/metabolism , Immunosuppressive Agents/pharmacology , Memory/physiology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/physiology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Humans
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.
Mol Neurobiol ; 58(7): 3043-3060, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33606195

ABSTRACT

The concept 'the retina as a window to the brain' has been increasingly explored in Alzheimer´s disease (AD) in recent years, since some patients present visual alterations before the first symptoms of dementia. The retina is an extension of the brain and can be assessed by noninvasive methods. However, assessing the retina for AD diagnosis is still a matter of debate. Using the triple transgenic mouse model of AD (3xTg-AD; males), this study was undertaken to investigate whether the retina and brain (hippocampus and cortex) undergo similar molecular and cellular changes during the early stages (4 and 8 months) of the pathology, and if the retina can anticipate the alterations occurring in the brain. We assessed amyloid-beta (Aß) and hyperphosphorylated tau (p-tau) levels, barrier integrity, cell death, neurotransmitter levels, and glial changes. Overall, the retina, hippocampus, and cortex of 3xTg-AD are not significantly affected at these early stages. However, we detected a few differential changes in the retina and brain regions, and particularly a different profile in microglia branching in the retina and hippocampus, only at 4 months, where the number and length of the processes decreased in the retina and increased in the hippocampus. In summary, at the early stages of pathology, the retina, hippocampus, and cortex are not significantly affected but already present some molecular and cellular alterations. The retina did not mirror the changes detected in the brain, and these observations should be taking into account when using the retina as a potential diagnostic tool for AD.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Brain/metabolism , Retina/metabolism , Alzheimer Disease/pathology , Animals , Brain/pathology , Cell Differentiation/physiology , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Organ Culture Techniques , Retina/pathology
20.
J Neurochem ; 112(1): 1-12, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19845827

ABSTRACT

Neurodegenerative diseases, generally characterized by a progressive deterioration in the structure and function of the brain, represent one of the world's major unsolved health problems. Therefore, it is urgent to discover therapeutic targets for the design of effective strategies for the treatment of these diseases. Recent findings demonstrated that the induction of the hypoxia signaling pathway with the concomitant stabilization and transcriptional activation of the transcription factor hypoxia-inducible factor 1 (HIF-1) could mediate neuroprotective events. It has been shown that HIF-1 triggers the expression of genes involved in oxygen transport, glycolytic metabolism, angiogenesis, cell survival, apoptosis, and others processes that can interfere with cell survival. Here, we discuss the current knowledge pertaining to the regulation of HIF signaling pathway. The potential neuroprotective role of HIF-1 induction in cerebral ischemic stroke and Alzheimer's, Parkinson's, and Huntington's diseases will be also discussed. The elucidation of the mechanisms involved in HIF-1-mediated neuroprotection could be important for the development of effective therapies to mitigate or prevent neurodegenerative diseases.


Subject(s)
Hypoxia-Inducible Factor 1/biosynthesis , Nerve Degeneration/metabolism , Animals , Drug Delivery Systems/trends , Humans , Hypoxia/metabolism , Hypoxia/prevention & control , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Nerve Degeneration/prevention & control , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/prevention & control , Neuroprotective Agents/administration & dosage , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL