Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 164.613
Filter
1.
PLoS One ; 19(5): e0303833, 2024.
Article in English | MEDLINE | ID: mdl-38768175

ABSTRACT

Fatigue can lead to several health issues and is particularly prevalent among elderly individuals with chronic inflammatory conditions. Ninjin'yoeito, a traditional Japanese herbal medicine, is used to address fatigue and malaise, anorexia, and anemia. This study aimed to examine whether relieving inflammation in the brain and skeletal muscle of senescence-accelerated mice prone 8 (SAMP8) could reduce fatigue-like conditions associated with aging. First, SAMP8 mice were divided into two groups, with and without ninjin'yoeito treatment. The ninjin'yoeito-treated group received a diet containing 3% ninjin'yoeito for a period of 4 months starting at 3 months of age. At 7 months of age, all mice underwent motor function, treadmill fatigue, and behavioral tests. They were then euthanized and the skeletal muscle weight, muscle cross-sectional area, and concentration of interleukin (IL)-1ß and IL-1 receptor antagonist (IL-1RA) in both the brain and skeletal muscle were measured. The results showed that the ninjin'yoeito-treated group had higher motor function and spontaneous locomotor activity than the untreated group did and ran for significantly longer in the treadmill fatigue test. Moreover, larger muscle cross-sectional area, lower IL-1ß concentrations, and higher IL-1RA concentrations were observed in both the brain and skeletal muscle tissues of the ninjin'yoeito-treated group than in the untreated group. The results suggest that ninjin'yoeito improves age-related inflammatory conditions in both the central and peripheral tissues and reduces fatigue.


Subject(s)
Aging , Brain , Drugs, Chinese Herbal , Fatigue , Inflammation , Muscle, Skeletal , Animals , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Aging/drug effects , Fatigue/drug therapy , Brain/drug effects , Brain/metabolism , Brain/pathology , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Male , Inflammation/drug therapy , Inflammation/pathology , Interleukin-1beta/metabolism
2.
Cell Mol Life Sci ; 81(1): 225, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38769116

ABSTRACT

Ischemic stroke induces neovascularization of the injured tissue as an attempt to promote structural repair and neurological recovery. Angiogenesis is regulated by pericytes that potently react to ischemic stroke stressors, ranging from death to dysfunction. Platelet-derived growth factor (PDGF) receptor (PDGFR)ß controls pericyte survival, migration, and interaction with brain endothelial cells. PDGF-D a specific ligand of PDGFRß is expressed in the brain, yet its regulation and role in ischemic stroke pathobiology remains unexplored. Using experimental ischemic stroke mouse model, we found that PDGF-D is transiently induced in brain endothelial cells at the injury site in the subacute phase. To investigate the biological significance of PDGF-D post-ischemic stroke regulation, its subacute expression was either downregulated using siRNA or upregulated using an active recombinant form. Attenuation of PDGF-D subacute induction exacerbates neuronal loss, impairs microvascular density, alters vascular permeability, and increases microvascular stalling. Increasing PDGF-D subacute bioavailability rescues neuronal survival and improves neurological recovery. PDGF-D subacute enhanced bioavailability promotes stable neovascularization of the injured tissue and improves brain perfusion. Notably, PDGF-D enhanced bioavailability improves pericyte association with brain endothelial cells. Cell-based assays using human brain pericyte and brain endothelial cells exposed to ischemia-like conditions were applied to investigate the underlying mechanisms. PDGF-D stimulation attenuates pericyte loss and fibrotic transition, while increasing the secretion of pro-angiogenic and vascular protective factors. Moreover, PDGF-D stimulates pericyte migration required for optimal endothelial coverage and promotes angiogenesis. Our study unravels new insights into PDGF-D contribution to neurovascular protection after ischemic stroke by rescuing the functions of pericytes.


Subject(s)
Endothelial Cells , Ischemic Stroke , Lymphokines , Pericytes , Platelet-Derived Growth Factor , Pericytes/metabolism , Pericytes/pathology , Animals , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Mice , Lymphokines/metabolism , Lymphokines/genetics , Platelet-Derived Growth Factor/metabolism , Humans , Endothelial Cells/metabolism , Male , Mice, Inbred C57BL , Brain/metabolism , Brain/pathology , Disease Models, Animal , Neovascularization, Physiologic , Cell Movement
3.
Cell Mol Life Sci ; 81(1): 224, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38769196

ABSTRACT

Synaptic loss is an early event in the penumbra area after an ischemic stroke. Promoting synaptic preservation in this area would likely improve functional neurological recovery. We aimed to detect proteins involved in endogenous protection mechanisms of synapses in the penumbra after stroke and to analyse potential beneficial effects of these candidates for a prospective stroke treatment. For this, we performed Liquid Chromatography coupled to Mass Spectrometry (LC-MS)-based proteomics of synaptosomes isolated from the ipsilateral hemispheres of mice subjected to experimental stroke at different time points (24 h, 4 and 7 days) and compared them to sham-operated mice. Proteomic analyses indicated that, among the differentially expressed proteins between the two groups, cystatin C (CysC) was significantly increased at 24 h and 4 days following stroke, before returning to steady-state levels at 7 days, thus indicating a potential transient and intrinsic rescue mechanism attempt of neurons. When CysC was applied to primary neuronal cultures subjected to an in vitro model of ischemic damage, this treatment significantly improved the preservation of synaptic structures. Notably, similar effects were observed when CysC was loaded into brain-derived extracellular vesicles (BDEVs). Finally, when CysC contained in BDEVs was administered intracerebroventricularly to stroked mice, it significantly increased the expression of synaptic markers such as SNAP25, Homer-1, and NCAM in the penumbra area compared to the group supplied with empty BDEVs. Thus, we show that CysC-loaded BDEVs promote synaptic protection after ischemic damage in vitro and in vivo, opening the possibility of a therapeutic use in stroke patients.


Subject(s)
Brain Ischemia , Brain , Cystatin C , Extracellular Vesicles , Mice, Inbred C57BL , Synapses , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Cystatin C/metabolism , Synapses/metabolism , Mice , Male , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain/metabolism , Brain/pathology , Proteomics/methods , Synaptosomes/metabolism , Neurons/metabolism , Stroke/metabolism , Stroke/pathology , Stroke/therapy , Cells, Cultured , Disease Models, Animal
4.
Gen Physiol Biophys ; 43(3): 263-271, 2024 May.
Article in English | MEDLINE | ID: mdl-38774925

ABSTRACT

Lithium (Li) is a mood-stabilizing drug. Although one of the potential mechanisms underlying the neuroprotective effects of lithium is related to its antioxidative effect, its mechanisms of action are not fully understood. Herein we aimed to investigate the impact of varied dosages of long-term lithium therapy on oxidative stress parameters in the brains of healthy rats, and on anxiety-like behaviors, and whether any changes in behavior can be attributed to modifications in oxidative stress levels within the brain. Thirty-two adult Wistar albino male rats were randomly assigned to four treatment groups. While the control (C) group was fed with a standard diet, low Li (1.4 g/kg/diet), moderate Li (1.8 g/kg/diet), and high Li (2.2 g/kg/diet) groups were fed with lithium bicarbonate (Li2CO3) for 30 days. Malondialdehyde increased, while superoxide dismutase and catalase levels decreased in the brains of the high Li group animals. In addition, anxiety-like behaviors of animals increased in the high Li group considering fewer entries to and less time spent in the open arms of the elevated plus maze test. Our findings underscore the potential adverse effects of prolonged lithium treatment, especially at doses approaching the upper therapeutic range. The induction of toxicity, manifested through heightened oxidative stress, appears to be a key mechanism contributing to the observed increase in anxiety-like behaviors. Consequently, caution is warranted when considering extended lithium therapy at higher doses, emphasizing the need for further research to delineate the precise mechanisms underlying these effects and to inform safer therapeutic practices.


Subject(s)
Anxiety , Brain , Dose-Response Relationship, Drug , Oxidative Stress , Rats, Wistar , Animals , Oxidative Stress/drug effects , Male , Rats , Anxiety/chemically induced , Anxiety/drug therapy , Brain/drug effects , Brain/metabolism , Lithium/pharmacology , Lithium/administration & dosage , Behavior, Animal/drug effects , Drug Administration Schedule , Lithium Compounds/pharmacology , Lithium Compounds/administration & dosage
5.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38775708

ABSTRACT

In utero infection and maternal inflammation can adversely impact fetal brain development. Maternal systemic illness, even in the absence of direct fetal brain infection, is associated with an increased risk of neuropsychiatric disorders in affected offspring. The cell types mediating the fetal brain response to maternal inflammation are largely unknown, hindering the development of novel treatment strategies. Here, we show that microglia, the resident phagocytes of the brain, highly express receptors for relevant pathogens and cytokines throughout embryonic development. Using a rodent maternal immune activation (MIA) model in which polyinosinic:polycytidylic acid is injected into pregnant mice, we demonstrate long-lasting transcriptional changes in fetal microglia that persist into postnatal life. We find that MIA induces widespread gene expression changes in neuronal and non-neuronal cells; importantly, these responses are abolished by selective genetic deletion of microglia, indicating that microglia are required for the transcriptional response of other cortical cell types to MIA. These findings demonstrate that microglia play a crucial durable role in the fetal response to maternal inflammation, and should be explored as potential therapeutic cell targets.


Subject(s)
Brain , Inflammation , Microglia , Poly I-C , Animals , Microglia/metabolism , Microglia/immunology , Female , Pregnancy , Mice , Brain/pathology , Brain/immunology , Brain/metabolism , Inflammation/pathology , Inflammation/genetics , Poly I-C/pharmacology , Fetus , Mice, Inbred C57BL , Gene Expression Regulation, Developmental , Neurons/metabolism
6.
J Mol Neurosci ; 74(2): 55, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38776015

ABSTRACT

The dysregulation of lipid metabolism has been strongly associated with Alzheimer's disease (AD) and has intricate connections with various aspects of disease progression, such as amyloidogenesis, bioenergetic deficit, oxidative stress, neuroinflammation, and myelin degeneration. Here, a comprehensive bioinformatic assessment was conducted on lipid metabolism genes in the brains and peripheral blood of AD-derived transcriptome datasets, characterizing the correlation between differentially expressed genes (DEGs) of lipid metabolism and disease pathologies, as well as immune cell preferences. Through the application of weighted gene co-expression network analysis (WGCNA), modules eigengenes related to lipid metabolism were pinpointed, and the examination of their molecular functions within biological processes, molecular pathways, and their associations with pathological phenotypes and molecular networks has been characterized. Analysis of biological networks indicates notable discrepancies in the expression patterns of the DEGs between neuronal and immune cells, as well as variations in cell type enrichments within both brain tissue and peripheral blood. Additionally, drugs targeting the DEGs from central and peripheral and a diagnostic model for hub genes from the blood were retrieved and assessed, some of which were shown to be useful for therapeutic and diagnostic. These results revealed the distinctive pattern of transcriptionally abnormal lipid metabolism in central, peripheral, and immune cell activation, providing valuable insight into lipid metabolism for diagnosing and guiding more effective treatment for AD.


Subject(s)
Alzheimer Disease , Lipid Metabolism , Transcriptome , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Humans , Lipid Metabolism/genetics , Brain/metabolism , Gene Regulatory Networks
7.
Mol Biol Rep ; 51(1): 572, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38722394

ABSTRACT

BACKGROUND: Alzheimer's disease is a leading neurological disorder that gradually impairs memory and cognitive abilities, ultimately leading to the inability to perform even basic daily tasks. Teriflunomide is known to preserve neuronal activity and protect mitochondria in the brain slices exposed to oxidative stress. The current research was undertaken to investigate the teriflunomide's cognitive rescuing abilities against scopolamine-induced comorbid cognitive impairment and its influence on phosphatidylinositol-3-kinase (PI3K) inhibition-mediated behavior alteration in mice. METHODS: Swiss albino mice were divided into 7 groups; vehicle control, scopolamine, donepezil + scopolamine, teriflunomide (10 mg/kg) + scopolamine; teriflunomide (20 mg/kg) + scopolamine, LY294002 and LY294002 + teriflunomide (20 mg/kg). Mice underwent a nine-day protocol, receiving scopolamine injections (2 mg/kg) for the final three days to induce cognitive impairment. Donepezil, teriflunomide, and LY294002 treatments were given continuously for 9 days. MWM, Y-maze, OFT and rota-rod tests were conducted on days 7 and 9. On the last day, blood samples were collected for serum TNF-α analysis, after which the mice were sacrificed, and brain samples were harvested for oxidative stress analysis. RESULTS: Scopolamine administration for three consecutive days increased the time required to reach the platform in the MWM test, whereas, reduced the percentage of spontaneous alternations in the Y-maze, number of square crossing in OFT and retention time in the rota-rod test. In biochemical analysis, scopolamine downregulated the brain GSH level, whereas it upregulated the brain TBARS and serum TNF-α levels. Teriflunomide treatment effectively mitigated all the behavioral and biochemical alterations induced by scopolamine. Furthermore, LY294002 administration reduced the memory function and GSH level, whereas, uplifted the serum TNF-α levels. Teriflunomide abrogated the memory-impairing, GSH-lowering, and TNF-α-increasing effects of LY294002. CONCLUSION: Our results delineate that the improvement in memory, locomotion, and motor coordination might be attributed to the oxidative and inflammatory stress inhibitory potential of teriflunomide. Moreover, PI3K inhibition-induced memory impairment might be attributed to reduced GSH levels and increased TNF-α levels.


Subject(s)
Cognitive Dysfunction , Crotonates , Hydroxybutyrates , Nitriles , Oxidative Stress , Toluidines , Animals , Nitriles/pharmacology , Mice , Hydroxybutyrates/pharmacology , Crotonates/pharmacology , Toluidines/pharmacology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Oxidative Stress/drug effects , Male , Disease Models, Animal , Maze Learning/drug effects , Behavior, Animal/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Scopolamine/pharmacology , Chromones/pharmacology , Memory/drug effects , Cognition/drug effects , Brain/metabolism , Brain/drug effects , Morpholines/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Donepezil/pharmacology
8.
Mol Biol Rep ; 51(1): 640, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38727848

ABSTRACT

Memory issues are a prevalent symptom in different neurodegenerative diseases and can also manifest in certain psychiatric conditions. Despite limited medications approved for treating memory problems, research suggests a lack of sufficient options in the market. Studies indicate that a significant percentage of elderly individuals experience various forms of memory disorders. Metformin, commonly prescribed for type 2 diabetes, has shown neuroprotective properties through diverse mechanisms. This study explores the potential of metformin in addressing memory impairments. The current research gathered its data by conducting an extensive search across electronic databases including PubMed, Web of Science, Scopus, and Google Scholar. Previous research suggests that metformin enhances brain cell survival and memory function in both animal and clinical models by reducing oxidative stress, inflammation, and cell death while increasing beneficial neurotrophic factors. The findings of the research revealed that metformin is an effective medication for enhancing various types of memory problems in numerous studies. Given the rising incidence of memory disorders, it is plausible to utilize metformin, which is an affordable and accessible drug. It is often recommended as a treatment to boost memory.


Subject(s)
Memory Disorders , Metformin , Metformin/therapeutic use , Metformin/pharmacology , Memory Disorders/drug therapy , Humans , Animals , Oxidative Stress/drug effects , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Memory/drug effects , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Brain/drug effects , Brain/metabolism
9.
Methods Mol Biol ; 2799: 139-150, 2024.
Article in English | MEDLINE | ID: mdl-38727906

ABSTRACT

Epilepsy is one of the most represented neurological diseases worldwide. However, in many cases, the precise molecular mechanisms of epileptogenesis and ictiogenesis are unknown. Because of their important role in synaptic function and neuronal excitability, NMDA receptors are implicated in various epileptogenic mechanisms. Most of these are subunit specific and require a precise analysis of the subunit composition of the NMDARs implicated. Here, we describe an express electrophysiological method to analyze the contribution of NMDAR subunits to spontaneous postsynaptic activity in identified cells in brain slices using patch clamp whole cell recordings.


Subject(s)
Patch-Clamp Techniques , Receptors, N-Methyl-D-Aspartate , Synapses , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Patch-Clamp Techniques/methods , Synapses/metabolism , Synapses/physiology , Brain/metabolism , Brain/cytology , Neurons/metabolism , Mice , Rats , Protein Subunits/metabolism
10.
Methods Mol Biol ; 2799: 243-255, 2024.
Article in English | MEDLINE | ID: mdl-38727911

ABSTRACT

Zebrafish are a powerful system to study brain development and to dissect the activity of complex circuits. One advantage is that they display complex behaviors, including prey capture, learning, responses to photic and acoustic stimuli, and social interaction (Dreosti et al., Front Neural Circuits 9:39, 2015; Bruckner et al., PLoS Biol 20:e3001838, 2022; Zoodsma et al., Mol Autism 13:38, 2022) that can be probed to assess brain function. Many of these behaviors are easily assayed at early larval stages, offering a noninvasive and high-throughput readout of nervous system function. Additionally, larval zebrafish readily uptake small molecules dissolved in water making them ideal for behavioral-based drug screens. Together, larval zebrafish and their behavioral repertoire offer a means to rapidly dissect brain circuitry and can serve as a template for high-throughput small molecule screens.NMDA receptor subunits are highly conserved in zebrafish compared to mammals (Zoodsma et al., Mol Autism 13:38, 2022; Cox et al., Dev Dyn 234:756-766, 2005; Zoodsma et al., J Neurosci 40:3631-3645, 2020). High amino acid and domain structure homology between humans and zebrafish underlie conserved functional similarities. Here we describe a set of behavioral assays that are useful to study the NMDA receptor activity in brain function.


Subject(s)
Behavior, Animal , Receptors, N-Methyl-D-Aspartate , Zebrafish , Animals , Zebrafish/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Behavior, Animal/drug effects , Larva/metabolism , Brain/metabolism , Brain/drug effects , High-Throughput Screening Assays/methods
11.
Front Endocrinol (Lausanne) ; 15: 1343759, 2024.
Article in English | MEDLINE | ID: mdl-38752176

ABSTRACT

Syndromic autism spectrum conditions (ASC), such as Klinefelter syndrome, also manifest hypogonadism. Compared to the popular Extreme Male Brain theory, the Enhanced Perceptual Functioning model explains the connection between ASC, savant traits, and giftedness more seamlessly, and their co-emergence with atypical sexual differentiation. Overexcitability of primary sensory inputs generates a relative enhancement of local to global processing of stimuli, hindering the abstraction of communication signals, in contrast to the extraordinary local information processing skills in some individuals. Weaker inhibitory function through gamma-aminobutyric acid type A (GABAA) receptors and the atypicality of synapse formation lead to this difference, and the formation of unique neural circuits that process external information. Additionally, deficiency in monitoring inner sensory information leads to alexithymia (inability to distinguish one's own emotions), which can be caused by hypoactivity of estrogen and oxytocin in the interoceptive neural circuits, comprising the anterior insular and cingulate gyri. These areas are also part of the Salience Network, which switches between the Central Executive Network for external tasks and the Default Mode Network for self-referential mind wandering. Exploring the possibility that estrogen deficiency since early development interrupts GABA shift, causing sensory processing atypicality, it helps to evaluate the co-occurrence of ASC with attention deficit hyperactivity disorder, dyslexia, and schizophrenia based on phenotypic and physiological bases. It also provides clues for understanding the common underpinnings of these neurodevelopmental disorders and gifted populations.


Subject(s)
Androgens , Autism Spectrum Disorder , Estrogens , Humans , Androgens/deficiency , Androgens/metabolism , Estrogens/metabolism , Estrogens/deficiency , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/physiopathology , Male , Sex Differentiation/physiology , Klinefelter Syndrome/physiopathology , Klinefelter Syndrome/metabolism , Perception/physiology , Brain/metabolism
12.
Proc Natl Acad Sci U S A ; 121(21): e2321496121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38753517

ABSTRACT

RNASET2-deficient leukodystrophy is a rare infantile white matter disorder mimicking a viral infection and resulting in severe psychomotor impairments. Despite its severity, there is little understanding of cellular mechanisms of pathogenesis and no treatments. Recent research using the rnaset2 mutant zebrafish model has suggested that microglia may be the drivers of the neuropathology, due to their failure to digest apoptotic debris during neurodevelopment. Therefore, we developed a strategy for microglial replacement through transplantation of adult whole kidney marrow-derived macrophages into embryonic hosts. Using live imaging, we revealed that transplant-derived macrophages can engraft within host brains and express microglia-specific markers, suggesting the adoption of a microglial phenotype. Tissue-clearing strategies revealed the persistence of transplanted cells in host brains beyond embryonic stages. We demonstrated that transplanted cells clear apoptotic cells within the brain, as well as rescue overactivation of the antiviral response otherwise seen in mutant larvae. RNA sequencing at the point of peak transplant-derived cell engraftment confirms that transplantation can reduce the brain-wide immune response and particularly, the antiviral response, in rnaset2-deficient brains. Crucially, this reduction in neuroinflammation resulted in behavioral rescue-restoring rnaset2 mutant motor activity to wild-type (WT) levels in embryonic and juvenile stages. Together, these findings demonstrate the role of microglia as the cellular drivers of neuropathology in rnaset2 mutants and that macrophage transplantation is a viable strategy for microglial replacement in the zebrafish. Therefore, microglia-targeted interventions may have therapeutic benefits in RNASET2-deficient leukodystrophy.


Subject(s)
Brain , Disease Models, Animal , Macrophages , Microglia , Zebrafish Proteins , Zebrafish , Animals , Microglia/metabolism , Microglia/pathology , Macrophages/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/metabolism , Brain/pathology , Brain/metabolism , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , Leukoencephalopathies/metabolism
13.
Gac Med Mex ; 160(1): 1-8, 2024.
Article in English | MEDLINE | ID: mdl-38753562

ABSTRACT

BACKGROUND: Protein interactions participate in many molecular mechanisms involved in cellular processes. The human TATA box binding protein (hTBP) interacts with Antennapedia (Antp) through its N-terminal region, specifically via its glutamine homopeptides. This PolyQ region acts as a binding site for other transcription factors under normal conditions, but when it expands, it generates spinocerebellar ataxia 17 (SCA17), whose protein aggregates in the brain prevent its correct functioning. OBJECTIVE: To determine whether the hTBP glutamine-rich region is involved in its interaction with homeoproteins and the role it plays in the formation of protein aggregates in SCA17. MATERIAL AND METHODS: We characterized hTBP interaction with other homeoproteins using BiFC, and modeled SCA17 in Drosophila melanogaster by targeting hTBPQ80 to the fly brain using UAS/GAL4. RESULTS: There was hTBP interaction with homeoproteins through its glutamine-rich region, and hTBP protein aggregates with expanded glutamines were found to affect the locomotor capacity of flies. CONCLUSIONS: The study of hTBP interactions opens the possibility for the search for new therapeutic strategies in neurodegenerative pathologies such as SCA17.


ANTECEDENTES: Las interacciones proteicas participan en una gran cantidad de mecanismos moleculares que rigen los procesos celulares. La proteína de unión a la caja TATA humana (hTBP) interacciona con Antennapedia (Antp) a través de su extremo N-terminal, específicamente a través de sus homopéptidos de glutaminas. Esta región PolyQ sirve como sitio de unión a factores de transcripción en condiciones normales, pero cuando se expande genera la ataxia espinal cerebelosa 17 (SCA17), cuyos agregados proteicos en el cerebro impiden su funcionamiento correcto. OBJETIVO: Determinar si la región rica en glutaminas de hTBP interviene en su interacción con homeoproteínas y el papel que tiene en la formación de agregados proteicos en SCA17. MATERIAL Y MÉTODOS: Se caracterizó la interacción de hTBP con otras homeoproteínas usando BiFC y se modeló SCA17 en Drosophila melanogaster dirigiendo hTBPQ80 al cerebro de las moscas usando UAS/GAL4. RESULTADOS: Existió interacción de hTBP con homeoproteínas a través de su región rica en glutaminas. Los agregados proteicos de hTBP con las glutaminas expandidas afectaron la capacidad locomotriz de las moscas. CONCLUSIONES: El estudio de las interacciones de hTBP abre la posibilidad para la búsqueda de nuevas estrategias terapéuticas en patologías neurodegenerativas como SCA17.


Subject(s)
Disease Models, Animal , Drosophila melanogaster , Spinocerebellar Ataxias , TATA-Box Binding Protein , Animals , Drosophila melanogaster/metabolism , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/genetics , TATA-Box Binding Protein/metabolism , TATA-Box Binding Protein/genetics , Humans , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Glutamine/metabolism , Protein Aggregates/physiology , Peptides/metabolism , Brain/metabolism
14.
Neurol Neuroimmunol Neuroinflamm ; 11(4): e200257, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38754047

ABSTRACT

OBJECTIVES: To assess whether the rate of change in synaptic proteins isolated from neuronally enriched extracellular vesicles (NEVs) is associated with brain and retinal atrophy in people with multiple sclerosis (MS). METHODS: People with MS were followed with serial blood draws, MRI (MRI), and optical coherence tomography (OCT) scans. NEVs were immunocaptured from plasma, and synaptopodin and synaptophysin proteins were measured using ELISA. Subject-specific rates of change in synaptic proteins, as well as brain and retinal atrophy, were determined and correlated. RESULTS: A total of 50 people with MS were included, 46 of whom had MRI and 45 had OCT serially. The rate of change in NEV synaptopodin was associated with whole brain (rho = 0.31; p = 0.04), cortical gray matter (rho = 0.34; p = 0.03), peripapillary retinal nerve fiber layer (rho = 0.37; p = 0.01), and ganglion cell/inner plexiform layer (rho = 0.41; p = 0.006) atrophy. The rate of change in NEV synaptophysin was also correlated with whole brain (rho = 0.31; p = 0.04) and cortical gray matter (rho = 0.31; p = 0.049) atrophy. DISCUSSION: NEV-derived synaptic proteins likely reflect neurodegeneration and may provide additional circulating biomarkers for disease progression in MS.


Subject(s)
Atrophy , Brain , Extracellular Vesicles , Multiple Sclerosis , Retina , Synaptophysin , Humans , Male , Female , Middle Aged , Extracellular Vesicles/metabolism , Adult , Brain/pathology , Brain/diagnostic imaging , Brain/metabolism , Retina/pathology , Retina/diagnostic imaging , Retina/metabolism , Multiple Sclerosis/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/diagnostic imaging , Synaptophysin/metabolism , Tomography, Optical Coherence , Magnetic Resonance Imaging , Microfilament Proteins/metabolism
15.
Sci Rep ; 14(1): 10669, 2024 05 09.
Article in English | MEDLINE | ID: mdl-38724577

ABSTRACT

Anaesthetics are used daily in human and veterinary medicine as well as in scientific research. Anaesthetics have an impact on cell homeostasis especially through modulation of protein post-translational modifications. O-GlcNAcylation, a ubiquitous post-translational modification, plays a role in many biological processes. The aims of this study were to evaluate whether (1) anaesthesia influences O-GlcNAcylation and (2) its stimulation affects physiological parameters. Male Wistar rats (n = 38) were anaesthetized with ketamine-xylazine or isoflurane. They randomly received either an intravenous injection of Ringer's lactate or NButGT (10mg/kg) in order to increase O-GlcNAcylation levels. One hour after induction of anaesthesia, haemodynamic parameters and plasmatic markers were evaluated. Heart, brain and lungs were harvested and O-GlcNAcylation levels and O-GlcNAc-related enzymes were evaluated by western blot. Cardiac and pulmonary O-GlcNAcylation levels and cardiac, cerebral and pulmonary O-GlcNAc associated enzyme expression were not impacted with anaesthesia. Compared with ketamine-xylazine, isoflurane had a lower impact on blood pressure, heart rate and glycaemia. Pharmacological stimulation of O-GlcNAcylation by NButGT did not affect the physiological parameters. This study offers unprecedented insights into the regulation of O-GlcNAcylation and O-GlcNAc related enzymes during anaesthesia. Pharmacological stimulation of O-GlcNAcylation over a 1-h period did not disrupt the physiological balance in healthy anaesthetized rats.


Subject(s)
Isoflurane , Ketamine , Rats, Wistar , Xylazine , Animals , Male , Rats , Isoflurane/pharmacology , Ketamine/pharmacology , Xylazine/pharmacology , Anesthesia , Acetylglucosamine/metabolism , Protein Processing, Post-Translational , Brain/metabolism , N-Acetylglucosaminyltransferases/metabolism , Heart Rate/drug effects , Lung/metabolism , Anesthetics/pharmacology , Blood Pressure/drug effects , Hemodynamics
16.
Sci Rep ; 14(1): 10709, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38729980

ABSTRACT

Three years after SARS-CoV-2 emerged as a global infectious threat, the virus has become endemic. The neurological complications such as depression, anxiety, and other CNS complications after COVID-19 disease are increasing. The brain, and CSF have been shown as viral reservoirs for SARS-CoV-2, yielding a potential hypothesis for CNS effects. Thus, we investigated the CNS pharmacology of orally dosed nirmatrelvir/ritonavir (NMR/RTV). Using both an in vitro and an in vivo rodent model, we investigated CNS penetration and potential pharmacodynamic activity of NMR. Through pharmacokinetic modeling, we estimated the median CSF penetration of NMR to be low at 18.11% of plasma with very low accumulation in rodent brain tissue. Based on the multiples of the 90% maximal effective concentration (EC90) for SARS-CoV-2, NMR concentrations in the CSF and brain do not achieve an exposure level similar to that of plasma. A median of only 16% of all the predicted CSF concentrations in rats were > 3xEC90 (unadjusted for protein binding). This may have implications for viral persistence and neurologic post-acute sequelae of COVID-19 if increased NMR penetration in the CNS leads to decreased CNS viral loads and decreased CNS inflammation.


Subject(s)
Leukocytes, Mononuclear , Ritonavir , SARS-CoV-2 , Animals , Rats , Ritonavir/pharmacokinetics , SARS-CoV-2/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Humans , Male , Brain/metabolism , Brain/virology , COVID-19 Drug Treatment , COVID-19/virology , COVID-19/cerebrospinal fluid , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Rats, Sprague-Dawley , Central Nervous System/metabolism , Central Nervous System/virology
17.
Int J Mol Sci ; 25(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38731864

ABSTRACT

The human brain possesses three predominate phospholipids, phosphatidylcholine (PC), phosphatidylethanolamine (PE) and phosphatidylserine (PS), which account for approximately 35-40%, 35-40%, and 20% of the brain's phospholipids, respectively. Mitochondrial membranes are relatively diverse, containing the aforementioned PC, PE, and PS, as well as phosphatidylinositol (PI) and phosphatidic acid (PA); however, cardiolipin (CL) and phosphatidylglycerol (PG) are exclusively present in mitochondrial membranes. These phospholipid interactions play an essential role in mitochondrial fusion and fission dynamics, leading to the maintenance of mitochondrial structural and signaling pathways. The essential nature of these phospholipids is demonstrated through the inability of mitochondria to tolerate alteration in these specific phospholipids, with changes leading to mitochondrial damage resulting in neural degeneration. This review will emphasize how the structure of phospholipids relates to their physiologic function, how their metabolism facilitates signaling, and the role of organ- and mitochondria-specific phospholipid compositions. Finally, we will discuss the effects of global ischemia and reperfusion on organ- and mitochondria-specific phospholipids alongside the novel therapeutics that may protect against injury.


Subject(s)
Brain , Heart Arrest , Mitochondria , Phospholipids , Humans , Phospholipids/metabolism , Mitochondria/metabolism , Animals , Brain/metabolism , Brain/pathology , Heart Arrest/metabolism , Signal Transduction , Mitochondrial Membranes/metabolism , Mitochondrial Dynamics
18.
PeerJ ; 12: e17342, 2024.
Article in English | MEDLINE | ID: mdl-38737745

ABSTRACT

Background: N-Ethylmaleimide (NEM), an agonist of the potassium chloride cotransporters 2 (KCC2) receptor, has been correlated with neurosuppressive outcomes, including decreased pain perception and the prevention of epileptic seizures. Nevertheless, its relationship with sleep-inducing effects remains unreported. Objective: The present study aimed to investigate the potential enhancement of NEM on the sleep-inducing properties of alprazolam (Alp). Methods: The test of the righting reflex was used to identify the appropriate concentrations of Alp and NEM for inducing sleep-promoting effects in mice. Total sleep duration and sleep quality were evaluated through EEG/EMG analysis. The neural mechanism underlying the sleep-promoting effect was examined through c-fos immunoreactivity in the brain using immunofluorescence. Furthermore, potential CNS-side effects of the combination Alp and NEM were assessed using LABORAS automated home-cage behavioral phenotyping. Results: Combination administration of Alp (1.84 mg/kg) and NEM (1.0 mg/kg) significantly decreased sleep latency and increased sleep duration in comparison to administering 1.84 mg/kg Alp alone. This effect was characterized by a notable increase in REM duration. The findings from c-fos immunoreactivity indicated that NEM significantly suppressed neuron activation in brain regions associated with wakefulness. Additionally, combination administration of Alp and NEM showed no effects on mouse neural behaviors during automated home cage monitoring. Conclusions: This study is the first to propose and demonstrate a combination therapy involving Alp and NEM that not only enhances the hypnotic effect but also mitigates potential CNS side effects, suggesting its potential application in treating insomnia.


Subject(s)
Alprazolam , Drug Synergism , Sleep , Animals , Alprazolam/pharmacology , Alprazolam/administration & dosage , Mice , Male , Sleep/drug effects , Electroencephalography/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Brain/drug effects , Brain/metabolism , Reflex, Righting/drug effects , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/administration & dosage
19.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(4): 303-310, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38710514

ABSTRACT

Objective To clarify the relationship between astrocyte activation patterns and disease progression in epidemic encephalitis B (Japanese encephalitis). Methods First, a mouse model of epidemic encephalitis B was constructed by foot-pad injection of Japanese encephalitis virus (JEV), and the expression of viral protein NS3 in different brain regions was detected by immunofluorescence assay (IFA). Next, IFA, RNA sequencing (RNA-seq) and real-time quantitative PCR (qRT-PCR) were used to clarify the changes in the astrocyte activation patterns at different stages of epidemic encephalitis B. Finally, intracerebroventricular administration of irisin was conducted to regulate the proportion of activation in complement C3-positive A1 astrocytes and S100A10-positive A2 astrocytes, investigating whether it could improve the body mass, behavioral scores, and brain tissue damage in a mouse model. Results NS3 protein was detected by IFA predominantly in the M1/M2 region of the motor cortex and the hippocampus. The number and volume of GFAP-positive astrocytes significantly increased in JEV-infected brain regions, in which the expression of multiple genes associated with A1/A2 astrocyte activation was significantly enhanced. Although intracerebroventricular or intraperitoneal injection of irisin did not improve the prognosis of epidemic encephalitis B, it inhibited the activation of A1 astrocytes and ameliorate neuroinflammation. Conclusion Neurons in the M1/M2 motor cortex and hippocampus are susceptible to JEV infection, in which the abnormal astrocyte activation contributes to the neuroinflammatory injury. Irisin administration may restrain A1 astrocyte activation and alleviate neuroinflammation following JEV infection.


Subject(s)
Astrocytes , Disease Models, Animal , Disease Progression , Encephalitis Virus, Japanese , Encephalitis, Japanese , Animals , Astrocytes/metabolism , Astrocytes/virology , Mice , Encephalitis, Japanese/immunology , Encephalitis Virus, Japanese/physiology , Brain/metabolism , Brain/virology , Brain/pathology , Male , Fibronectins/metabolism , Fibronectins/genetics
20.
AAPS PharmSciTech ; 25(5): 96, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710855

ABSTRACT

Central nervous system-related disorders have become a continuing threat to human life and the current statistic indicates an increasing trend of such disorders worldwide. The primary therapeutic challenge, despite the availability of therapies for these disorders, is to sustain the drug's effective concentration in the brain while limiting its accumulation in non-targeted areas. This is attributed to the presence of the blood-brain barrier and first-pass metabolism which limits the transportation of drugs to the brain irrespective of popular and conventional routes of drug administration. Therefore, there is a demand to practice alternative routes for predictable drug delivery using advanced drug delivery carriers to overcome the said obstacles. Recent research attracted attention to intranasal-to-brain drug delivery for promising targeting therapeutics in the brain. This review emphasizes the mechanisms to deliver therapeutics via different pathways for nose-to-brain drug delivery with recent advancements in delivery and formulation aspects. Concurrently, for the benefit of future studies, the difficulties in administering medications by intranasal pathway have also been highlighted.


Subject(s)
Administration, Intranasal , Blood-Brain Barrier , Brain , Drug Delivery Systems , Administration, Intranasal/methods , Humans , Drug Delivery Systems/methods , Brain/metabolism , Blood-Brain Barrier/metabolism , Animals , Drug Carriers/chemistry , Pharmaceutical Preparations/administration & dosage , Nasal Mucosa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...