Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 208
Filter
Add more filters

Publication year range
1.
Acta Pharmacol Sin ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684799

ABSTRACT

Epilepsy is a chronic and debilitating neurological disorder, known for the occurrence of spontaneous and recurrent seizures. Despite the availability of antiseizure drugs, 30% of people with epilepsy experience uncontrolled seizures and drug resistance, evidencing that new therapeutic options are required. The process of epileptogenesis involves the development and expansion of tissue capable of generating spontaneous recurrent seizures, during which numerous events take place, namely blood-brain barrier (BBB) dysfunction, and neuroinflammation. The consequent cerebrovascular dysfunction results in a lower seizure threshold, seizure recurrence, and chronic epilepsy. This suggests that improving cerebrovascular health may interrupt the pathological cycle responsible for disease development and progression. Krüppel-like factors (KLFs) are a family of zinc-finger transcription factors, encountered in brain endothelial cells, glial cells, and neurons. KLFs are known to regulate vascular function and changes in their expression are associated with neuroinflammation and human diseases, including epilepsy. Hence, KLFs have demonstrated various roles in cerebrovascular dysfunction and epileptogenesis. This review critically discusses the purpose of KLFs in epileptogenic mechanisms and BBB dysfunction, as well as the potential of their pharmacological modulation as therapeutic approach for epilepsy treatment.

2.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39000036

ABSTRACT

Air pollution, a growing concern for public health, has been linked to various respiratory and cardiovascular diseases. Emerging evidence also suggests a link between exposure to air pollutants and neurodegenerative diseases, particularly Alzheimer's disease (AD). This review explores the composition and sources of air pollutants, including particulate matter, gases, persistent organic pollutants, and heavy metals. The pathophysiology of AD is briefly discussed, highlighting the role of beta-amyloid plaques, neurofibrillary tangles, and genetic factors. This article also examines how air pollutants reach the brain and exert their detrimental effects, delving into the neurotoxicity of air pollutants. The molecular mechanisms linking air pollution to neurodegeneration are explored in detail, focusing on oxidative stress, neuroinflammation, and protein aggregation. Preclinical studies, including in vitro experiments and animal models, provide evidence for the direct effects of pollutants on neuronal cells, glial cells, and the blood-brain barrier. Epidemiological studies have reported associations between exposure to air pollution and an increased risk of AD and cognitive decline. The growing body of evidence supporting air pollution as a modifiable risk factor for AD underscores the importance of considering environmental factors in the etiology and progression of neurodegenerative diseases, in the face of worsening global air quality.


Subject(s)
Air Pollutants , Air Pollution , Alzheimer Disease , Humans , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Air Pollution/adverse effects , Air Pollutants/adverse effects , Air Pollutants/toxicity , Risk Factors , Animals , Particulate Matter/adverse effects , Oxidative Stress , Neurodegenerative Diseases/etiology , Environmental Exposure/adverse effects , Brain/pathology , Brain/metabolism
3.
Int J Mol Sci ; 24(18)2023 Sep 16.
Article in English | MEDLINE | ID: mdl-37762479

ABSTRACT

Licochalcone A (Lico-A) is a flavonoid compound derived from the root of the Glycyrrhiza species, a plant commonly used in traditional Chinese medicine. While the Glycyrrhiza species has shown promise in treating various diseases such as cancer, obesity, and skin diseases due to its active compounds, the investigation of Licochalcone A's effects on the central nervous system and its potential application in Alzheimer's disease (AD) treatment have garnered significant interest. Studies have reported the neuroprotective effects of Lico-A, suggesting its potential as a multitarget compound. Lico-A acts as a PTP1B inhibitor, enhancing cognitive activity through the BDNF-TrkB pathway and exhibiting inhibitory effects on microglia activation, which enables mitigation of neuroinflammation. Moreover, Lico-A inhibits c-Jun N-terminal kinase 1, a key enzyme involved in tau phosphorylation, and modulates the brain insulin receptor, which plays a role in cognitive processes. Lico-A also acts as an acetylcholinesterase inhibitor, leading to increased levels of the neurotransmitter acetylcholine (Ach) in the brain. This mechanism enhances cognitive capacity in individuals with AD. Finally, Lico-A has shown the ability to reduce amyloid plaques, a hallmark of AD, and exhibits antioxidant properties by activating the nuclear factor erythroid 2-related factor 2 (Nrf2), a key regulator of antioxidant defense mechanisms. In the present review, we discuss the available findings analyzing the potential of Lico-A as a neuroprotective agent. Continued research on Lico-A holds promise for the development of novel treatments for cognitive disorders and neurodegenerative diseases, including AD. Further investigations into its multitarget action and elucidation of underlying mechanisms will contribute to our understanding of its therapeutic potential.


Subject(s)
Alzheimer Disease , Chalcones , Humans , Antioxidants/pharmacology , Antioxidants/therapeutic use , Alzheimer Disease/drug therapy , Acetylcholinesterase , Chalcones/pharmacology , Chalcones/therapeutic use
4.
Mol Med ; 28(1): 48, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35508978

ABSTRACT

BACKGROUND AND AIM: The appearance of alterations in normal metabolic activity has been increasingly considered a risk factor for the development of sporadic and late-onset neurodegenerative diseases. In this report, we induced chronic metabolic stress by feeding of a high-fat diet (HFD) in order to study its consequences in cognition. We also studied the effects of a loss of function of isoforms 1 and 3 of the c-Jun N-terminal Kinases (JNK), stress and cell death response elements. METHODS: Animals were fed either with conventional chow or with HFD, from their weaning until their sacrifice at 9 months. Before sacrifice, body weight, intraperitoneal glucose and insulin tolerance test (IP-GTT and IP­ITT) were performed to evaluate peripheral biometrics. Additionally, cognitive behavioral tests and analysis of spine density were performed to assess cognitive function. Molecular studies were carried out to confirm the effects of metabolic stressors in the hippocampus relative to cognitive loss. RESULTS: Our studies demonstrated that HFD in Jnk3-/- lead to synergetic responses. Loss of function of JNK3 led to increased body weight, especially when exposed to an HFD and they had significantly decreased response to insulin. These mice also showed increased stress in the endoplasmic reticulum and diminished cognitive capacity. However, loss of function of JNK1 promoted normal or heightened energetic metabolism and preserved cognitive function even when chronically metabolically stressed. CONCLUSIONS: Downregulation of JNK3 does not seem to be a suitable target for the modulation of energetic-cognitive dysregulations while loss of function of JNK1 seems to promote a good metabolic-cognitive profile, just like resistance to the negative effects of chronic feeding with HFD.


Subject(s)
Hippocampus , Mitogen-Activated Protein Kinase 8 , Animals , Body Weight , Cognition , Diet, High-Fat/adverse effects , Hippocampus/metabolism , Insulin/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism
5.
Nutr Neurosci ; 25(12): 2627-2637, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34789070

ABSTRACT

Deregulations like the loss of sensitivity to insulin (insulin resistance) and chronic inflammation are alterations very commonly found in sporadic forms of neurodegenerative pathologies. Thus, finding strategies to protect against them, may lead to a reduction in the incidence and/or affectation of these pathologies. The grape seed-derived proanthocyanidins extract (GSPE) is a mixture of compounds highly enriched in polyphenols and flavonoids that have shown to have a wide range of therapeutic benefits due to their antioxidant and anti-inflammatory properties. OBJECTIVES: This study aimed to assess the protective effects of a short pre-treatment of GSPE in the hippocampus against a prolonged feeding with cafeteria diet. METHODS: GSPE was administered for 10 days followed by 12 weeks of cafeteria diet. We analyzed transcriptional activity of genes and protein expression of key mediators of neurodegeneration in brain samples. RESULTS: Results indicated that GSPE was able to protect against cellular damage through the activation of AKT, as well as promote the maintenance of mitochondrial function by conserving the OXPHOS complexes and upregulating the antioxidant SOD. DISCUSSION: We observed that GSPE decreased inflammatory activation as observed through the downregulation of JNK, IL6 and TNFα, just like the reduction in reactive profile of astrocytes. Overall, the data presented here offers an interesting and hopeful initial step for future long-term studies on the beneficial effects of a supplementation of common diets with polyphenol and flavonoid substances for the amelioration of typical early hallmarks of neurodegeneration.


Subject(s)
Proanthocyanidins , Rats , Animals , Proanthocyanidins/pharmacology , Antioxidants/pharmacology , Rats, Wistar , Diet , Polyphenols/pharmacology , Hippocampus , Mitochondria
6.
Int J Mol Sci ; 22(17)2021 Sep 02.
Article in English | MEDLINE | ID: mdl-34502457

ABSTRACT

(1) Background: The c-Jun-NH2-terminal protein kinase (JNK) is a mitogen-activated protein kinase involved in regulating physiological processes in the central nervous system. However, the dual genetic deletion of Mkk4 and Mkk7 (upstream activators of JNK) in adult mice is not reported. The aim of this study was to induce the genetic deletion of Mkk4/Mkk7 in adult mice and analyze their effect in hippocampal neurogenesis. (2) Methods: To achieve this goal, Actin-CreERT2 (Cre+/-), Mkk4flox/flox, Mkk7flox/flox mice were created. The administration of tamoxifen in these 2-month-old mice induced the gene deletion (Actin-CreERT2 (Cre+/-), Mkk4∆/∆, Mkk7∆/∆ genotype), which was verified by PCR, Western blot, and immunohistochemistry techniques. (3) Results: The levels of MKK4/MKK7 at 7 and 14 days after tamoxifen administration were not eliminated totally in CNS, unlike what happens in the liver and heart. These data could be correlated with the high levels of these proteins in CNS. In the hippocampus, the deletion of Mkk4/Mkk7 induced a misalignment position of immature hippocampal neurons together with alterations in their dendritic architecture pattern and maturation process jointly to the diminution of JNK phosphorylation. (4) Conclusion: All these data supported that the MKK4/MKK7-JNK pathway has a role in adult neurogenic activity.


Subject(s)
Hippocampus/physiology , MAP Kinase Kinase 4/physiology , MAP Kinase Kinase 7/physiology , MAP Kinase Signaling System , Neurogenesis , Animals , Doublecortin Protein , Gene Deletion , Mice, Transgenic
7.
J Neuroinflammation ; 17(1): 22, 2020 Jan 14.
Article in English | MEDLINE | ID: mdl-31937343

ABSTRACT

BACKGROUND: The increasing risk of obesity and diabetes among other metabolic disorders are the consequence of shifts in dietary patterns with high caloric-content food intake. We previously reported that megalin regulates energy homeostasis using blood-brain barrier (BBB) endothelial megalin-deficient (EMD) mice, since these animals developed obesity and metabolic syndrome upon normal chow diet administration. Obesity in mid-life appears to be related to greater dementia risk and represents an increasing global health issue. We demonstrated that EMD phenotype induced impaired learning ability and recognition memory, neurodegeneration, neuroinflammation, reduced neurogenesis, and mitochondrial deregulation associated with higher mitochondrial mass in cortical tissues. METHODS: EMD mice were subjected to normal chow and high-fat diet (HFD) for 14 weeks and metabolic changes were evaluated. RESULTS: Surprisingly, BBB megalin deficiency protected against HFD-induced obesity improving glucose tolerance and preventing hepatic steatosis. Compared to wild type (wt), the brain cortex in EMD mice showed increased levels of the mitochondrial biogenesis regulator, peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), and uncoupling protein 2 (UCP2), a thermogenic protein involved in the regulation of energy metabolism. This agreed with the previously found increased mitochondrial mass in the transgenic mice. Upon HFD challenge, we demonstrated these two proteins were found elevated in wt mice but reported no changes over the already increased levels in EMD animals. CONCLUSION: We propose a protective role for megalin on diet-induce obesity, suggesting this could be related to metabolic disturbances found in dementia through brain endocrine system communications.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Diet, High-Fat/adverse effects , Endothelium, Vascular/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Animals , Energy Metabolism/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism
8.
J Neurochem ; 149(2): 255-268, 2019 04.
Article in English | MEDLINE | ID: mdl-30734928

ABSTRACT

Insulin resistance has negative consequences on the physiological functioning of the nervous system. The appearance of type 3 diabetes in the brain leads to the development of the sporadic form of Alzheimer's disease. The c-Jun N-terminal kinases (JNK), a subfamily of the Mitogen Activated Protein Kinases, are enzymes composed by three different isoforms with differential modulatory activity against the insulin receptor (IR) and its substrate. This research focused on understanding the regulatory role of JNK2 on the IR, as well as study the effect of a high-fat diet (HFD) in the brain. Our observations determined how JNK2 ablation did not induce compensatory responses in the expression of the other isoforms but led to an increase in JNKs total activity. HFD-fed animals also showed an increased activity profile of the JNKs. These animals also displayed endoplasmic reticulum stress and up-regulation of the protein tyrosine phosphatase 1B (PTP1B) and the suppressor of cytokine signalling 3 protein. Consequently, a reduction in insulin sensitivity was detected and it is correlated with a decrease on the signalling of the IR. Moreover, cognitive impairment was observed in all groups but only wild-type genotype animals fed with HFD showed neuroinflammatory responses. In conclusion, HFD and JNK2 absence cause alterations in normal cognitive activity by altering the signalling of the IR. These affectations are related to the appearance of endoplasmic reticulum stress and an increase in the levels of inhibitory proteins like PTP1B and suppressor of cytokine signalling 3 protein. Cover Image for this issue: doi: 10.1111/jnc.14502.


Subject(s)
Brain/metabolism , Cognition/physiology , Diet, High-Fat/adverse effects , Mitogen-Activated Protein Kinase 9/metabolism , Receptor, Insulin/metabolism , Animals , Disease Models, Animal , Endoplasmic Reticulum Stress/physiology , Insulin Resistance/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
Pharmacol Res ; 145: 104255, 2019 07.
Article in English | MEDLINE | ID: mdl-31075308

ABSTRACT

After decades of research, the molecular neuropathology of Alzheimer's disease (AD) is still one of the hot topics in biomedical sciences. Some studies suggest that soluble amyloid ß (Aß) oligomers act as causative agents in the development of AD and could be initiators of its complex neurodegenerative cascade. On the other hand, there is also evidence pointing to Aß oligomers as mere aggravators, with an arguable role in the origin of the disease. In this line of research, the relative contribution of soluble Aß oligomers to neuronal damage associated with metabolic disorders such as Type 2 Diabetes Mellitus (T2DM) and obesity is being actively investigated. Some authors have proposed the endoplasmic reticulum (ER) stress and the induction of the unfolded protein response (UPR) as important mechanisms leading to an increase in Aß production and the activation of neuroinflammatory processes. Following this line of thought, these mechanisms could also cause cognitive impairment. The present review summarizes the current understanding on the neuropathological role of Aß associated with metabolic alterations induced by an obesogenic high fat diet (HFD) intake. It is believed that the combination of these two elements has a synergic effect, leading to the impairement of ER and mitochondrial functions, glial reactivity status alteration and inhibition of insulin receptor (IR) signalling. All these metabolic alterations would favour neuronal malfunction and, eventually, neuronal death by apoptosis, hence causing cognitive impairment and laying the foundations for late-onset AD (LOAD). Moreover, since drugs enhancing the activation of cerebral insulin pathway can constitute a suitable strategy for the prevention of AD, we also discuss the scope of therapeutic approaches such as intranasal administration of insulin in clinical trials with AD patients.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Animals , Ceramides/metabolism , Cognitive Dysfunction/etiology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum Stress , Humans , Obesity/complications
10.
Int J Mol Sci ; 21(1)2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31905931

ABSTRACT

Previous studies have reported that the regulatory function of the different c-Jun N-terminal kinases isoforms (JNK1, JNK2, and JNK3) play an essential role in neurological disorders, such as epilepsy and metabolic-cognitive alterations. Accordingly, JNKs have emerged as suitable therapeutic strategies. In fact, it has been demonstrated that some unspecific JNK inhibitors exert antidiabetic and neuroprotective effects, albeit they usually show high toxicity or lack therapeutic value. In this sense, natural specific JNK inhibitors, such as Licochalcone A, are promising candidates. Nonetheless, research on the understanding of the role of each of the JNKs remains mandatory in order to progress on the identification of new selective JNK isoform inhibitors. In the present review, a summary on the current gathered data on the role of JNKs in pathology is presented, as well as a discussion on their potential role in pathologies like epilepsy and metabolic-cognitive injury. Moreover, data on the effects of synthetic small molecule inhibitors that modulate JNK-dependent pathways in the brain and peripheral tissues is reviewed.


Subject(s)
Cognitive Dysfunction/metabolism , Epilepsy/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Animals , Brain/metabolism , Cognitive Dysfunction/pathology , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum Stress , Epilepsy/pathology , Humans , Hypoglycemic Agents/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/drug effects , MAP Kinase Signaling System/physiology , Mitochondria/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neuroprotective Agents/antagonists & inhibitors , Protein Isoforms
11.
J Nanobiotechnology ; 16(1): 32, 2018 Mar 27.
Article in English | MEDLINE | ID: mdl-29587747

ABSTRACT

BACKGROUND: Memantine, drug approved for moderate to severe Alzheimer's disease, has not shown to be fully effective. In order to solve this issue, polylactic-co-glycolic (PLGA) nanoparticles could be a suitable solution to increase drug's action on the target site as well as decrease adverse effects. For these reason, Memantine was loaded in biodegradable PLGA nanoparticles, produced by double emulsion method and surface-coated with polyethylene glycol. MEM-PEG-PLGA nanoparticles (NPs) were aimed to target the blood-brain barrier (BBB) upon oral administration for the treatment of Alzheimer's disease. RESULTS: The production parameters were optimized by design of experiments. MEM-PEG-PLGA NPs showed a mean particle size below 200 nm (152.6 ± 0.5 nm), monomodal size distribution (polydispersity index, PI < 0.1) and negative surface charge (- 22.4 mV). Physicochemical characterization of NPs confirmed that the crystalline drug was dispersed inside the PLGA matrix. MEM-PEG-PLGA NPs were found to be non-cytotoxic on brain cell lines (bEnd.3 and astrocytes). Memantine followed a slower release profile from the NPs against the free drug solution, allowing to reduce drug administration frequency in vivo. Nanoparticles were able to cross BBB both in vitro and in vivo. Behavioral tests carried out on transgenic APPswe/PS1dE9 mice demonstrated to enhance the benefit of decreasing memory impairment when using MEM-PEG-PLGA NPs in comparison to the free drug solution. Histological studies confirmed that MEM-PEG-PLGA NPs reduced ß-amyloid plaques and the associated inflammation characteristic of Alzheimer's disease. CONCLUSIONS: Memantine NPs were suitable for Alzheimer's disease and more effective than the free drug.


Subject(s)
Alzheimer Disease/drug therapy , Antiparkinson Agents/pharmacokinetics , Cognitive Dysfunction/drug therapy , Drug Carriers , Memantine/pharmacokinetics , Nanoparticles/chemistry , Plaque, Amyloid/drug therapy , Administration, Oral , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Antiparkinson Agents/chemistry , Antiparkinson Agents/pharmacology , Astrocytes/cytology , Astrocytes/drug effects , Blood-Brain Barrier/metabolism , Cell Line , Cell Survival/drug effects , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Drug Compounding/methods , Emulsions , Humans , Male , Maze Learning/drug effects , Memantine/chemistry , Memantine/pharmacology , Mice , Mice, Transgenic , Neurons/cytology , Neurons/drug effects , Particle Size , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Polyesters/chemistry , Polyethylene Glycols/chemistry
12.
Nanomedicine ; 14(4): 1073-1085, 2018 06.
Article in English | MEDLINE | ID: mdl-29454994

ABSTRACT

Temporal lobe epilepsy is the most common type of pharmacoresistant epilepsy in adults. Epigallocatechin-3-gallate has aroused much interest because of its multiple therapeutic effects, but its instability compromises the potential effectiveness. PEGylated-PLGA nanoparticles of Epigallocatechin-3-gallate were designed to protect the drug and to increase the brain delivery. Nanoparticles were prepared by the double emulsion method and cytotoxicity, behavioral, Fluoro-Jade C, Iba1 and GFAP immunohistochemistry studies were carried out to determine their effectiveness. Nanoparticles showed an average size of 169 nm, monodisperse population, negative surface charge, encapsulation efficiency of 95% and sustained release profile. Cytotoxicity assays exhibited that these nanocarriers were non-toxic. Behavioral test showed that nanoparticles reduced most than free drug the number of epileptic episodes and their intensity. Neurotoxicity and immunohistochemistry studies confirmed a decrease in neuronal death and neuroinflammation. In conclusion, Epigallocatechin-3-gallate PEGylated-PLGA nanoparticles could be a suitable strategy for the treatment of temporal lobe epilepsy.


Subject(s)
Catechin/analogs & derivatives , Epilepsy, Temporal Lobe/drug therapy , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Animals , Catechin/chemistry , Catechin/therapeutic use , Drug Carriers/chemistry , Emulsions , Epilepsy, Temporal Lobe/metabolism , Mice , Mice, Inbred C57BL , PC12 Cells , Particle Size , Rats , Seizures/drug therapy
13.
J Neuroinflammation ; 14(1): 26, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28143489

ABSTRACT

BACKGROUND: The multiligand receptor megalin controls the brain uptake of a number of ligands, including insulin and leptin. Despite the role of megalin in the transport of these metabolically relevant hormones, the role of megalin at the blood-brain-barrier (BBB) has not yet been explored in the context of metabolic regulation. METHODS: Here we investigate the role of brain endothelial megalin in energy metabolism and leptin signaling using an endothelial cell-specific megalin deficient (EMD) mouse model. RESULTS: We found megalin is important to protect mice from developing obesity and metabolic syndrome when mice are fed a normal chow diet. EMD mice developed neuroinflammation, by triggering several pro-inflammatory cytokines, displayed reduced neurogenesis and mitochondrial deregulation. CONCLUSIONS: These results implicate brain endothelial megalin expression in obesity-related metabolic changes through the leptin signaling pathway proposing a potential link between obesity and neurodegeneration.


Subject(s)
Encephalitis/genetics , Encephalitis/pathology , Endothelial Cells/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Obesity/genetics , Obesity/pathology , Age Factors , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/physiopathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Doublecortin Domain Proteins , Eating/genetics , Glial Fibrillary Acidic Protein/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Mitochondria/pathology , Mitochondria/ultrastructure , Neuropeptides/metabolism , Phosphopyruvate Hydratase/metabolism , STAT3 Transcription Factor/metabolism
14.
J Biomed Sci ; 24(1): 27, 2017 May 09.
Article in English | MEDLINE | ID: mdl-28486943

ABSTRACT

BACKGROUND: Neonatal monosodium glutamate (MSG) treatment triggers excitotoxicity and induces a degenerative process that affects several brain regions in a way that could lead to epileptogenesis. Na+/Ca2+ exchangers (NCX1-3) are implicated in Ca2+ brain homeostasis; normally, they extrude Ca2+ to control cell inflammation, but after damage and in epilepsy, they introduce Ca2+ by acting in the reverse mode, amplifying the damage. Changes in NCX3 expression in the hippocampus have been reported immediately after neonatal MSG treatment. In this study, the expression level of NCX1-3 in the entorhinal cortex (EC) and hippocampus (Hp); and the effects of blockade of NCXs on the seizures induced by 4-Aminopyridine (4-AP) were analysed in adult rats after neonatal MSG treatment. KB-R7943 was applied as NCXs blocker, but is more selective to NCX3 in reverse mode. METHODS: Neonatal MSG treatment was applied to newborn male rats at postnatal days (PD) 1, 3, 5, and 7 (4 g/kg of body weight, s.c.). Western blot analysis was performed on total protein extracts from the EC and Hp to estimate the expression level of NCX1-3 proteins in relative way to the expression of ß-actin, as constitutive protein. Electrographic activity of the EC and Hp were acquired before and after intracerebroventricular (i.c.v.) infusion of 4-AP (3 nmol) and KB-R7943 (62.5 pmol), alone or in combination. All experiments were performed at PD60. Behavioural alterations were also recorder. RESULTS: Neonatal MSG treatment significantly increased the expression of NCX3 protein in both studied regions, and NCX1 protein only in the EC. The 4-AP-induced epileptiform activity was significantly higher in MSG-treated rats than in controls, and KB-R7943 co-administered with 4-AP reduced the epileptiform activity in more prominent way in MSG-treated rats than in controls. CONCLUSIONS: The long-term effects of neonatal MSG treatment include increases on functional expression of NCXs (mainly of NCX3) in the EC and Hp, which seems to contribute to improve the control that KB-R7943 exerted on the seizures induced by 4-AP in adulthood. The results obtained here suggest that the blockade of NCXs could improve seizure control after an excitotoxic process; however, this must be better studied.


Subject(s)
4-Aminopyridine/toxicity , Anticonvulsants/pharmacology , Sodium Glutamate/toxicity , Thiourea/analogs & derivatives , Animals , Entorhinal Cortex/drug effects , Entorhinal Cortex/metabolism , Entorhinal Cortex/physiopathology , Gene Expression/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Homeodomain Proteins/metabolism , Infusions, Intraventricular , Male , Rats , Rats, Wistar , Thiourea/pharmacology
15.
Nanomedicine ; 13(3): 1171-1182, 2017 04.
Article in English | MEDLINE | ID: mdl-27986603

ABSTRACT

Dexibuprofen loaded pegylated poly(lactic-co-glycolic) nanospheres prepared by solvent diffusion method were designed to increase Dexibuprofen brain delivery reducing systemic side effects. Nanospheres exhibited a mean particle size around 200 nm (195.4 nm), monomodal population and negative surface charge. Drug loaded nanospheres showed a sustained release profile, allowing to modify the posology in vivo. Nanospheres were non-toxic neither in brain endothelial cells nor astrocytes and do not cause blood-brain barrier disruption. Nanospheres were able to partially cross the cells barrier and release the drug after co-culture in vitro experiments, increasing Dexibuprofen permeation coefficient. Behavioral tests performed in APPswe/PS1dE9 mice (mice model of familial Alzheimer's disease) showed that nanospheres reduce memory impairment more efficiently than the free drug. Developed nanospheres decrease brain inflammation leading to ß-amyloid plaques reduction. According to these results, chronical oral Dexibuprofen pegylated poly(lactic-co-glycolic) nanosystems could constitute a suitable strategy for the prevention of neurodegeneration.


Subject(s)
Alzheimer Disease/prevention & control , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Drug Carriers/chemistry , Ibuprofen/analogs & derivatives , Nanospheres/chemistry , Polyethylene Glycols/chemistry , Polyglactin 910/chemistry , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Cell Line , Coculture Techniques , Disease Models, Animal , Ibuprofen/administration & dosage , Ibuprofen/pharmacokinetics , Ibuprofen/therapeutic use , Male , Memory/drug effects , Mice , Mice, Inbred C57BL
16.
Biochim Biophys Acta ; 1852(9): 1687-99, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26003667

ABSTRACT

Global obesity is a pandemic status, estimated to affect over 2 billion people, that has resulted in an enormous strain on healthcare systems worldwide. The situation is compounded by the fact that apart from the direct costs associated with overweight pathology, obesity presents itself with a number of comorbidities, including an increased risk for the development of neurodegenerative disorders. Alzheimer disease (AD), the main cause of senile dementia, is no exception. Spectacular failure of the pharmaceutical industry to come up with effective AD treatment strategies is forcing the broader scientific community to rethink the underlying molecular mechanisms leading to cognitive decline. To this end, the emphasis is once again placed on the experimental animal models of the disease. In the current study, we have focused on the effects of a high-fat diet (HFD) on hippocampal-dependent memory in C57/Bl6 Wild-type (WT) and APPswe/PS1dE9 (APP/PS1) mice, a well-established mouse model of familial AD. Our results indicate that the continuous HFD administration starting at the time of weaning is sufficient to produce ß-amyloid-independent, hippocampal-dependent memory deficits measured by a 2-object novel-object recognition test (NOR) in mice as early as 6months of age. Furthermore, the resulting metabolic syndrome appears to have direct effects on brain insulin regulation and mitochondrial function. We have observed pathological changes related to both the proximal and distal insulin signaling pathway in the brains of HFD-fed WT and APP/PS1 mice. These changes are accompanied by a significantly reduced OXPHOS metabolism, suggesting that mitochondria play an important role in hippocampus-dependent memory formation and retention in both the HFD-treated and AD-like rodents at a relatively young age.

17.
Neural Plast ; 2016: 8501693, 2016.
Article in English | MEDLINE | ID: mdl-26881137

ABSTRACT

Alzheimer's disease (AD) currently presents one of the biggest healthcare issues in the developed countries. There is no effective treatment capable of slowing down disease progression. In recent years the main focus of research on novel pharmacotherapies was based on the amyloidogenic hypothesis of AD, which posits that the beta amyloid (Aß) peptide is chiefly responsible for cognitive impairment and neuronal death. The goal of such treatments is (a) to reduce Aß production through the inhibition of ß and γ secretase enzymes and (b) to promote dissolution of existing cerebral Aß plaques. However, this approach has proven to be only modestly effective. Recent studies suggest an alternative strategy centred on the inhibition of the downstream Aß signalling, particularly at the synapse. Aß oligomers may cause aberrant N-methyl-D-aspartate receptor (NMDAR) activation postsynaptically by forming complexes with the cell-surface prion protein (PrPC). PrPC is enriched at the neuronal postsynaptic density, where it interacts with Fyn tyrosine kinase. Fyn activation occurs when Aß is bound to PrPC-Fyn complex. Fyn causes tyrosine phosphorylation of the NR2B subunit of metabotropic glutamate receptor 5 (mGluR5). Fyn kinase blockers masitinib and saracatinib have proven to be efficacious in treating AD symptoms in experimental mouse models of the disease.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Animals , Benzamides , Benzodioxoles/administration & dosage , Benzodioxoles/therapeutic use , Brain/drug effects , Disease Models, Animal , Humans , Mice , Neurons/metabolism , Piperidines , PrPC Proteins/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/metabolism , Pyridines , Quinazolines/administration & dosage , Quinazolines/therapeutic use , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Serotonin/metabolism , Synapses/metabolism , Thiazoles/administration & dosage , Thiazoles/therapeutic use
18.
Biochim Biophys Acta ; 1842(9): 1556-66, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24887203

ABSTRACT

The present study had focused on the behavioral phenotype and gene expression profile of molecules related to insulin receptor signaling in the hippocampus of 3 and 6 month-old APPswe/PS1dE9 (APP/PS1) transgenic mouse model of Alzheimer's disease (AD). Elevated levels of the insoluble Aß (1-42) were detected in the brain extracts of the transgenic animals as early as 3 months of age, prior to the Aß plaque formation (pre-plaque stage). By the early plaque stage (6 months) both the soluble and insoluble Aß (1-40) and Aß (1-42) peptides were detectable. We studied the expression of genes related to memory function (Arc, Fos), insulin signaling, including insulin receptor (Insr), Irs1 and Irs2, as well as genes involved in insulin growth factor pathways, such as Igf1, Igf2, Igfr and Igfbp2. We also examined the expression and protein levels of key molecules related to energy metabolism (PGC1-α, and AMPK) and mitochondrial functionality (OXPHOS, TFAM, NRF1 and NRF2). 6 month-old APP/PS1 mice demonstrated impaired cognitive ability, were glucose intolerant and showed a significant reduction in hippocampal Insr and Irs2 transcripts. Further observations also suggest alterations in key cellular energy sensors that regulate the activities of a number of metabolic enzymes through phosphorylation, such as a decrease in the Prkaa2 mRNA levels and in the pAMPK (Thr172)/Total APMK ratio. Moreover, mRNA and protein analysis reveals a significant downregulation of genes essential for mitochondrial replication and respiratory function, including PGC-1α in hippocampal extracts of APP/PS1 mice, compared to age-matched wild-type controls at 3 and 6 months of age. Overall, the findings of this study show early alterations in genes involved in insulin and energy metabolism pathways in an APP/PS1 model of AD. These changes affect the activity of key molecules like NRF1 and PGC-1α, which are involved in mitochondrial biogenesis. Our results reinforce the hypothesis that the impairments in both insulin signaling and energy metabolism precede the development of AD amyloidogenesis.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/physiology , Disease Models, Animal , Energy Metabolism , Hippocampus/metabolism , Mitochondria/metabolism , Presenilin-1/physiology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloidogenic Proteins , Animals , Biomarkers/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Hippocampus/pathology , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
19.
Mol Pharm ; 12(11): 4137-45, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26440359

ABSTRACT

Receptor-mediated transcytosis has been widely studied as a possible strategy to transport neurotherapeutics across the blood-brain barrier (BBB). Monoclonal antibodies directed against the transferrin receptor (TfR) have been proposed as potential carrier candidates. A better understanding of the mechanisms involved in their cellular uptake and intracellular trafficking is required and could critically contribute to the improvement of delivery methods. Accordingly, we studied here the trafficking of gold nanoparticles (AuNPs) coated with the 8D3 anti-transferrin receptor antibody at the mouse BBB. 8D3-AuNPs were intravenously administered to mice and allowed to recirculate for a range of times, from 10 min to 24 h, before brain extraction and analysis by transmission electron microscope techniques. Our results indicated a TfR-mediated and clathrin-dependent internalization process by which 8D3-AuNPs internalize individually in vesicles. These vesicles then follow at least two different routes. On one hand, most vesicles enter intracellular processes of vesicular fusion and rearrangement in which the AuNPs end up accumulating in late endosomes, multivesicular bodies or lysosomes, which present a high AuNP content. On the other hand, a small percentage of the vesicles follow a different route in which they fuse with the abluminal membrane and open to the basal membrane. In these cases, the 8D3-AuNPs remain attached to the abluminal membrane, which suggests an endosomal escape, but not dissociation from TfR. Altogether, although receptor-mediated transport continues to be one of the most promising strategies to overcome the BBB, different optimization approaches need to be developed for efficient drug delivery.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Blood-Brain Barrier , Brain/metabolism , Drug Delivery Systems , Gold/chemistry , Metal Nanoparticles/chemistry , Receptors, Transferrin/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Male , Mice , Mice, Inbred ICR , Protein Transport , Tissue Distribution
20.
Biochim Biophys Acta ; 1832(8): 1241-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23507144

ABSTRACT

The role of peroxisome proliferator activator receptor (PPAR)ß/δ in the pathogenesis of Alzheimer's disease has only recently been explored through the use of PPARß/δ agonists. Here we evaluated the effects of PPARß/δ deficiency on the amyloidogenic pathway and tau hyperphosphorylation. PPARß/δ-null mice showed cognitive impairment in the object recognition task, accompanied by enhanced DNA-binding activity of NF-κB in the cortex and increased expression of IL-6. In addition, two NF-κB-target genes involved in ß-amyloid (Aß) synthesis and deposition, the ß site APP cleaving enzyme 1 (Bace1) and the receptor for advanced glycation endproducts (Rage), respectively, increased in PPARß/δ-null mice compared to wild type animals. The protein levels of glial fibrillary acidic protein (GFAP) increased in the cortex of PPARß/δ-null mice, which would suggest the presence of astrogliosis. Finally, tau hyperphosphorylation at Ser199 and enhanced levels of PHF-tau were associated with increased levels of the tau kinases CDK5 and phospho-ERK1/2 in the cortex of PPARß/δ(-/-) mice. Collectively, our findings indicate that PPARß/δ deficiency results in cognitive impairment associated with enhanced inflammation, astrogliosis and tau hyperphosphorylation in the cortex.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Cerebral Cortex/metabolism , PPAR-beta/deficiency , Receptors, Immunologic/metabolism , tau Proteins/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Cognition/physiology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Glial Fibrillary Acidic Protein , Inflammation , Interleukin-6/genetics , Interleukin-6/metabolism , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , PPAR-beta/genetics , PPAR-beta/metabolism , Phosphorylation , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , tau Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL