Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.163
Filter
1.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891789

ABSTRACT

This review addresses the role of tight junction proteins at the blood-brain barrier (BBB). Their expression is described, and their role in physiological and pathological processes at the BBB is discussed. Based on this, new approaches are depicted for paracellular drug delivery and diagnostics in the treatment of cerebral diseases. Recent data provide convincing evidence that, in addition to its impairment in the course of diseases, the BBB could be involved in the aetiology of CNS disorders. Further progress will be expected based on new insights in tight junction protein structure and in their involvement in signalling pathways.


Subject(s)
Blood-Brain Barrier , Tight Junction Proteins , Tight Junctions , Blood-Brain Barrier/metabolism , Humans , Tight Junction Proteins/metabolism , Animals , Tight Junctions/metabolism , Central Nervous System Diseases/metabolism , Signal Transduction
2.
Int J Mol Sci ; 25(12)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38928312

ABSTRACT

Serum amyloid A (SAA) proteins are highly conserved lipoproteins that are notoriously involved in the acute phase response and systemic amyloidosis, but their biological functions are incompletely understood. Recent work has shown that SAA proteins can enter the brain by crossing the intact blood-brain barrier (BBB), and that they can impair BBB functions. Once in the central nervous system (CNS), SAA proteins can have both protective and harmful effects, which have important implications for CNS disease. In this review of the thematic series on SAA, we discuss the existing literature that relates SAA to neuroinflammation and CNS disease, and the possible roles of the BBB in these relations.


Subject(s)
Blood-Brain Barrier , Serum Amyloid A Protein , Blood-Brain Barrier/metabolism , Humans , Serum Amyloid A Protein/metabolism , Animals , Central Nervous System Diseases/metabolism
3.
Int J Mol Sci ; 25(11)2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38892334

ABSTRACT

Noncoding RNAs (ncRNAs) are a class of nucleotide sequences that cannot be translated into peptides. ncRNAs can function post-transcriptionally by splicing complementary sequences of mRNAs or other ncRNAs or by directly engaging in protein interactions. Over the past few decades, the pervasiveness of ncRNAs in cell physiology and their pivotal roles in various diseases have been identified. One target regulated by ncRNAs is connexin (Cx), a protein that forms gap junctions and hemichannels and facilitates intercellular molecule exchange. The aberrant expression and misdistribution of connexins have been implicated in central nervous system diseases, cardiovascular diseases, bone diseases, and cancer. Current databases and technologies have enabled researchers to identify the direct or indirect relationships between ncRNAs and connexins, thereby elucidating their correlation with diseases. In this review, we selected the literature published in the past five years concerning disorders regulated by ncRNAs via corresponding connexins. Among it, microRNAs that regulate the expression of Cx43 play a crucial role in disease development and are predominantly reviewed. The distinctive perspective of the ncRNA-Cx axis interprets pathology in an epigenetic manner and is expected to motivate research for the development of biomarkers and therapeutics.


Subject(s)
Connexins , RNA, Untranslated , Humans , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Animals , Connexins/metabolism , Connexins/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Connexin 43/genetics , Connexin 43/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Gene Expression Regulation , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/therapy , Gap Junctions/metabolism , Gap Junctions/genetics , Central Nervous System Diseases/genetics , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/therapy
4.
Biomed Pharmacother ; 176: 116811, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38795641

ABSTRACT

Central nervous system (CNS) disorders exhibit exceedingly intricate pathogenic mechanisms. Pragmatic and effective solutions remain elusive, significantly compromising human life and health. Activating transcription factor 4 (ATF4) participates in the regulation of multiple pathophysiological processes, including CNS disorders. Considering the widespread involvement of ATF4 in the pathological process of CNS disorders, the targeted regulation of ATF4 by plant-derived bioactive compounds (PDBCs) may become a viable strategy for the treatment of CNS disorders. However, the regulatory relationship between PDBCs and ATF4 remains incompletely understood. Here, we aimed to comprehensively review the studies on PDBCs targeting ATF4 to ameliorate CNS disorders, thereby offering novel directions and insights for the treatment of CNS disorders. A computerized search was conducted on PubMed, Embase, Web of Science, and Google Scholar databases to identify preclinical experiments related to PDBCs targeting ATF4 for the treatment of CNS disorders. The search timeframe was from the inception of the databases to December 2023. Two assessors conducted searches using the keywords "ATF4," "Central Nervous System," "Neurological," "Alzheimer's disease," "Parkinson's Disease," "Stroke," "Spinal Cord Injury," "Glioblastoma," "Traumatic Brain Injury," and "Spinal Cord Injury." Overall, 31 studies were included, encompassing assessments of 27 PDBCs. Combining results from in vivo and in vitro studies, we observed that these PDBCs, via ATF4 modulation, prevent the deposition of amyloid-like fibers such as Aß, tau, and α-synuclein. They regulate ERS, reduce the release of inflammatory factors, restore mitochondrial membrane integrity to prevent oxidative stress, regulate synaptic plasticity, modulate autophagy, and engage anti-apoptotic mechanisms. Consequently, they exert neuroprotective effects in CNS disorders. Numerous PDBCs targeting ATF4 have shown potential in facilitating the restoration of CNS functionality, thereby presenting expansive prospects for the treatment of such disorders. However, future endeavors necessitate high-quality, large-scale, and comprehensive preclinical and clinical studies to further validate this therapeutic potential.


Subject(s)
Activating Transcription Factor 4 , Central Nervous System Diseases , Activating Transcription Factor 4/metabolism , Humans , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/metabolism , Animals , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
5.
Cells ; 13(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38727275

ABSTRACT

ATP-binding cassette (ABC) transporters play a crucial role for the efflux of a wide range of substrates across different cellular membranes. In the central nervous system (CNS), ABC transporters have recently gathered significant attention due to their pivotal involvement in brain physiology and neurodegenerative disorders, such as Alzheimer's disease (AD). Glial cells are fundamental for normal CNS function and engage with several ABC transporters in different ways. Here, we specifically highlight ABC transporters involved in the maintenance of brain homeostasis and their implications in its metabolic regulation. We also show new aspects related to ABC transporter function found in less recognized diseases, such as Huntington's disease (HD) and experimental autoimmune encephalomyelitis (EAE), as a model for multiple sclerosis (MS). Understanding both their impact on the physiological regulation of the CNS and their roles in brain diseases holds promise for uncovering new therapeutic options. Further investigations and preclinical studies are warranted to elucidate the complex interplay between glial ABC transporters and physiological brain functions, potentially leading to effective therapeutic interventions also for rare CNS disorders.


Subject(s)
ATP-Binding Cassette Transporters , Central Nervous System , Neuroglia , Humans , ATP-Binding Cassette Transporters/metabolism , Neuroglia/metabolism , Animals , Central Nervous System/metabolism , Central Nervous System/pathology , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/pathology
6.
Cell Mol Neurobiol ; 44(1): 46, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38743119

ABSTRACT

Central nervous system (CNS) disorders represent the leading cause of disability and the second leading cause of death worldwide, and impose a substantial economic burden on society. In recent years, emerging evidence has found that beta2 -microglobulin (B2M), a subunit of major histocompatibility complex class I (MHC-I) molecules, plays a crucial role in the development and progression in certain CNS diseases. On the one hand, intracellular B2M was abnormally upregulated in brain tumors and regulated tumor microenvironments and progression. On the other hand, soluble B2M was also elevated and involved in pathological stages in CNS diseases. Targeted B2M therapy has shown promising outcomes in specific CNS diseases. In this review, we provide a comprehensive summary and discussion of recent advances in understanding the pathological processes involving B2M in CNS diseases (e.g., Alzheimer's disease, aging, stroke, HIV-related dementia, glioma, and primary central nervous system lymphoma).


Subject(s)
Central Nervous System Diseases , beta 2-Microglobulin , Humans , beta 2-Microglobulin/metabolism , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/pathology , Animals
7.
Curr Opin Biotechnol ; 87: 103131, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38599012

ABSTRACT

Glial cells are important in maintaining homeostasis for neurons in the central nervous system (CNS). During CNS disease or after injury, glia react to altered microenvironments and often acquire altered functions that contribute to disease pathology. A major focus for research is utilizing stem cell (SC)-derived glia as a potential renewable source for cell replacement to restore function, including neuronal support, and as a model for disease states to identify therapeutic targets. In this review, we focus on SC differentiation protocols for deriving three types of glial cells, astrocytes, oligodendrocytes, and microglia. These SC-derived glia can be used to identify critical cues that contribute to CNS disease progression and aid in investigation of therapeutic targets.


Subject(s)
Central Nervous System Diseases , Neuroglia , Humans , Neuroglia/metabolism , Central Nervous System Diseases/therapy , Central Nervous System Diseases/metabolism , Animals , Cell Differentiation , Stem Cells/cytology , Cell Engineering/methods
8.
Neuropathol Appl Neurobiol ; 50(2): e12980, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38647003

ABSTRACT

Neuroinflammation, blood-brain barrier (BBB) dysfunction, neuron and glia injury/death and myelin damage are common central nervous system (CNS) pathologies observed in various neurological diseases and injuries. Serine protease inhibitor (Serpin) clade A member 3n (Serpina3n), and its human orthologue SERPINA3, is an acute-phase inflammatory glycoprotein secreted primarily by the liver into the bloodstream in response to systemic inflammation. Clinically, SERPINA3 is dysregulated in brain cells, cerebrospinal fluid and plasma in various neurological conditions. Although it has been widely accepted that Serpina3n/SERPINA3 is a reliable biomarker of reactive astrocytes in diseased CNS, recent data have challenged this well-cited concept, suggesting instead that oligodendrocytes and neurons are the primary sources of Serpina3n/SERPINA3. The debate continues regarding whether Serpina3n/SERPINA3 induction represents a pathogenic or a protective mechanism. Here, we propose possible interpretations for previously controversial data and present perspectives regarding the potential role of Serpina3n/SERPINA3 in CNS pathologies, including demyelinating disorders where oligodendrocytes are the primary targets. We hypothesise that the 'good' or 'bad' aspects of Serpina3n/SERPINA3 depend on its cellular sources, its subcellular distribution (or mis-localisation) and/or disease/injury types. Furthermore, circulating Serpina3n/SERPINA3 may cross the BBB to impact CNS pathologies. Cell-specific genetic tools are critically important to tease out the potential roles of cell type-dependent Serpina3n in CNS diseases/injuries.


Subject(s)
Serpins , Humans , Serpins/metabolism , Serpins/genetics , Animals , Central Nervous System Diseases/pathology , Central Nervous System Diseases/metabolism , Central Nervous System/pathology , Central Nervous System/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/metabolism
9.
J Mol Med (Berl) ; 102(6): 751-760, 2024 06.
Article in English | MEDLINE | ID: mdl-38639785

ABSTRACT

Annexins, a group of Ca2+-dependent phospholipid-binding proteins, exert diverse roles in neuronal development, normal central nervous system (CNS) functioning, neurological disorders, and CNS tumors. This paper reviews the roles of individual annexins (A1-A13) in these contexts. Annexins possess unique structural and functional features, such as Ca2+-dependent binding to phospholipids, participating in membrane organization, and modulating cell signaling. They are implicated in various CNS processes, including endocytosis, exocytosis, and stabilization of plasma membranes. Annexins exhibit dynamic roles in neuronal development, influencing differentiation, proliferation, and synaptic formation in CNS tissues. Notably, annexins such as ANXA1 and ANXA2 play roles in apoptosis and blood-brain barrier (BBB) integrity. Neurological disorders, including Alzheimer's disease, multiple sclerosis, and depression, involve annexin dysregulation, influencing neuroinflammation, blood-brain barrier integrity, and stress responses. Moreover, annexins contribute to the pathogenesis of CNS tumors, either promoting or suppressing tumor growth, angiogenesis, and invasion. Annexin expression patterns vary across different CNS tumor types, providing potential prognostic markers and therapeutic targets. This review underscores the multifaceted roles of annexins in the CNS, highlighting their importance in normal functioning, disease progression, and potential therapeutic interventions.


Subject(s)
Annexins , Central Nervous System , Humans , Annexins/metabolism , Animals , Central Nervous System/metabolism , Blood-Brain Barrier/metabolism , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/pathology , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology
10.
CNS Neurosci Ther ; 30(3): e14677, 2024 03.
Article in English | MEDLINE | ID: mdl-38497529

ABSTRACT

BACKGROUND: Exosomes are vesicles secreted by all types of mammalian cells. They are characterized by a double-layered lipid membrane structure. They serve as carriers for a plethora of signal molecules, including DNA, RNA, proteins, and lipids. Their unique capability of effortlessly crossing the blood-brain barrier underscores their critical role in the progression of various neurological disorders. This includes, but is not limited to, diseases such as Alzheimer's, Parkinson's, and ischemic stroke. Establishing stable and mature methods for isolating exosomes is a prerequisite for the study of exosomes and their biomedical significance. The extraction technologies of exosomes include differential centrifugation, density gradient centrifugation, size exclusion chromatography, ultrafiltration, polymer coprecipitation, immunoaffinity capture, microfluidic, and so forth. Each extraction technology has its own advantages and disadvantages, and the extraction standards of exosomes have not been unified internationally. AIMS: This review aimed to showcase the recent advancements in exosome isolation techniques and thoroughly compare the advantages and disadvantages of different methods. Furthermore, the significant research progress made in using exosomes for diagnosing and treating central nervous system (CNS) diseases has been emphasized. CONCLUSION: The varying isolation methods result in differences in the concentration, purity, and size of exosomes. The efficient separation of exosomes facilitates their widespread application, particularly in the diagnosis and treatment of CNS diseases.


Subject(s)
Central Nervous System Diseases , Exosomes , Humans , Exosomes/metabolism , Proteins/metabolism , Central Nervous System Diseases/therapy , Central Nervous System Diseases/metabolism
11.
Methods Mol Biol ; 2761: 1-26, 2024.
Article in English | MEDLINE | ID: mdl-38427225

ABSTRACT

Detection of merely apoptosis does not reveal the type of central nervous system (CNS) cells that are dying in the CNS diseases and injuries. In situ detection and estimation of amount of apoptosis specifically in neurons or glial cells (astrocytes, oligodendrocytes, and microglia) can unveil valuable information for designing therapeutics for protection of the CNS cells and functional recovery. A method was first developed and reported from our laboratory for in situ detection and estimation of amount of apoptosis precisely in neurons and glial cells using in vitro and in vivo models of CNS diseases and injuries. This is a combination of terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and double immunofluorescent labeling (DIFL) or simply TUNEL-n-DIFL method for in situ detection and estimation of amount of apoptosis in a specific CNS cell type. An anti-digoxigenin (DIG) IgG antibody conjugated with 7-amino-4-methylcoumarin-3-acetic acid (AMCA) for blue fluorescence, fluorescein isothiocyanate (FITC) for green fluorescence, or Texas Red (TR) for red fluorescence can be used for in situ detection of apoptotic cell DNA, which is earlier labeled with TUNEL using alkali-stable DIG-11-dUTP. A primary anti-NeuN (neurons), anti-GFAP (astrocytes), anti-MBP (oligodendrocytes), or anti-OX-42 (microglia) IgG antibody and a secondary IgG antibody conjugated with one of the above fluorophores (other than that of ani-DIG antibody) are used for in situ detection of apoptosis in a specific CNS cell type in the mixed culture and animal models of the CNS diseases and injuries.


Subject(s)
Apoptosis , Central Nervous System Diseases , Animals , In Situ Nick-End Labeling , Apoptosis/genetics , Neuroglia , Neurons/metabolism , Central Nervous System Diseases/metabolism , Disease Models, Animal , Immunoglobulin G/metabolism
12.
Cells ; 13(5)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38474374

ABSTRACT

Mitochondria, the energy suppliers of the cells, play a central role in a variety of cellular processes essential for survival or leading to cell death. Consequently, mitochondrial dysfunction is implicated in numerous general and CNS disorders. The clinical manifestations of mitochondrial dysfunction include metabolic disorders, dysfunction of the immune system, tumorigenesis, and neuronal and behavioral abnormalities. In this review, we focus on the mitochondrial role in the CNS, which has unique characteristics and is therefore highly dependent on the mitochondria. First, we review the role of mitochondria in neuronal development, synaptogenesis, plasticity, and behavior as well as their adaptation to the intricate connections between the different cell types in the brain. Then, we review the sparse knowledge of the mechanisms of exogenous mitochondrial uptake and describe attempts to determine their half-life and transplantation long-term effects on neuronal sprouting, cellular proteome, and behavior. We further discuss the potential of mitochondrial transplantation to serve as a tool to study the causal link between mitochondria and neuronal activity and behavior. Next, we describe mitochondrial transplantation's therapeutic potential in various CNS disorders. Finally, we discuss the basic and reverse-translation challenges of this approach that currently hinder the clinical use of mitochondrial transplantation.


Subject(s)
Central Nervous System Diseases , Mitochondrial Diseases , Humans , Mitochondria/metabolism , Central Nervous System/metabolism , Brain/metabolism , Central Nervous System Diseases/metabolism , Mitochondrial Diseases/metabolism
13.
J Allergy Clin Immunol ; 153(4): 913-923, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38365015

ABSTRACT

The immune system classically consists of 2 lines of defense, innate and adaptive, both of which interact with one another effectively to protect us against any pathogenic threats. Importantly, there is a diverse subset of cells known as innate-like T cells that act as a bridge between the innate and adaptive immune systems and are pivotal players in eliciting inflammatory immune responses. A growing body of evidence has demonstrated the regulatory impact of these innate-like T cells in central nervous system (CNS) diseases and that such immune cells can traffic into the brain in multiple pathological conditions, which can be typically attributed to the breakdown of the blood-brain barrier. However, until now, it has been poorly understood whether innate-like T cells have direct protective or causative properties, particularly in CNS diseases. Therefore, in this review, our attention is focused on discussing the critical roles of 3 unique subsets of unconventional T cells, namely, natural killer T cells, γδ T cells, and mucosal-associated invariant T cells, in the context of CNS diseases, disorders, and injuries and how the interplay of these immune cells modulates CNS pathology, in an attempt to gain a better understanding of their complex functions.


Subject(s)
Central Nervous System Diseases , Mucosal-Associated Invariant T Cells , Natural Killer T-Cells , Humans , Central Nervous System Diseases/metabolism , Immunity, Innate
14.
Article in English | MEDLINE | ID: mdl-38316554

ABSTRACT

In addition to their many functions in the healthy central nervous system (CNS), astrocytes respond to CNS damage and disease through a process called "reactivity." Recent evidence reveals that astrocyte reactivity is a heterogeneous spectrum of potential changes that occur in a context-specific manner. These changes are determined by diverse signaling events and vary not only with the nature and severity of different CNS insults but also with location in the CNS, genetic predispositions, age, and potentially also with "molecular memory" of previous reactivity events. Astrocyte reactivity can be associated with both essential beneficial functions as well as with harmful effects. The available information is rapidly expanding and much has been learned about molecular diversity of astrocyte reactivity. Emerging functional associations point toward central roles for astrocyte reactivity in determining the outcome in CNS disorders.


Subject(s)
Astrocytes , Central Nervous System Diseases , Astrocytes/metabolism , Humans , Central Nervous System Diseases/metabolism , Animals , Central Nervous System/metabolism , Signal Transduction
15.
Eur J Med Res ; 29(1): 15, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38173021

ABSTRACT

Early diagnosis and pharmacological treatment of central nervous system (CNS) diseases has been a long-standing challenge for clinical research due to the presence of the blood-brain barrier. Specific proteins and RNAs in brain-derived extracellular vesicles (EVs) usually reflect the corresponding state of brain disease, and therefore, EVs can be used as diagnostic biomarkers for CNS diseases. In addition, EVs can be engineered and fused to target cells for delivery of cargo, demonstrating the great potential of EVs as a nanocarrier platform. We review the progress of EVs as markers and drug carriers in the diagnosis and treatment of neurological diseases. The main areas include visual imaging, biomarker diagnosis and drug loading therapy for different types of CNS diseases. It is hoped that increased knowledge of EVs will facilitate their clinical translation in CNS diseases.


Subject(s)
Central Nervous System Diseases , Extracellular Vesicles , Humans , Brain , Extracellular Vesicles/metabolism , Blood-Brain Barrier , Biomarkers/metabolism , Central Nervous System Diseases/diagnosis , Central Nervous System Diseases/therapy , Central Nervous System Diseases/metabolism
16.
Psychiatry Clin Neurosci ; 78(2): 83-96, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37877617

ABSTRACT

Extracellular vesicles (EVs) are membrane-enclosed nanovesicles secreted by cells into the extracellular space and contain functional biomolecules, e.g. signaling receptors, bioactive lipids, nucleic acids, and proteins, which can serve as biomarkers. Neurons and glial cells secrete EVs, contributing to various physiological and pathological aspects of brain diseases. EVs confer their role in the bidirectional crosstalk between the central nervous system (CNS) and the periphery owing to their distinctive ability to cross the unique blood-brain barrier (BBB). Thus, EVs in the blood, cerebrospinal fluid (CSF), and urine can be intriguing biomarkers, enabling the minimally invasive diagnosis of CNS diseases. Although there has been an enormous interest in evaluating EVs as promising biomarkers, the lack of ultra-sensitive approaches for isolating and detecting brain-derived EVs (BDEVs) has hindered the development of efficient biomarkers. This review presents the recent salient findings of exosomal biomarkers, focusing on brain disorders. We summarize highly sensitive sensors for EV detection and state-of-the-art methods for single EV detection. Finally, the prospect of developing advanced EV analysis approaches for the non-invasive diagnosis of brain diseases is presented.


Subject(s)
Brain Diseases , Central Nervous System Diseases , Extracellular Vesicles , Humans , Brain/metabolism , Extracellular Vesicles/metabolism , Central Nervous System Diseases/diagnosis , Central Nervous System Diseases/metabolism , Brain Diseases/diagnosis , Biomarkers
17.
Adv Healthc Mater ; 13(12): e2304180, 2024 05.
Article in English | MEDLINE | ID: mdl-38112345

ABSTRACT

Microglia play a pivotal role in the central nervous system (CNS) homeostasis, acting as housekeepers and defenders of the surrounding environment. These cells can elicit their functions by shifting into two main phenotypes: pro-inflammatory classical phenotype, M1, and anti-inflammatory alternative phenotype, M2. Despite their pivotal role in CNS homeostasis, microglia phenotypes can influence the development and progression of several CNS disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, ischemic stroke, traumatic brain injuries, and even brain cancer. It is thus clear that the possibility of modulating microglia activation has gained attention as a therapeutic tool against many CNS pathologies. Nanomaterials are an unprecedented tool for manipulating microglia responses, in particular, to specifically target microglia and elicit an in situ immunomodulation activity. This review focuses the discussion on two main aspects: analyzing the possibility of using nanomaterials to stimulate a pro-inflammatory response of microglia against brain cancer and introducing nanostructures able to foster an anti-inflammatory response for treating neurodegenerative disorders. The final aim is to stimulate the analysis of the development of new microglia nano-immunomodulators, paving the way for innovative and effective therapeutic approaches for the treatment of CNS disorders.


Subject(s)
Central Nervous System Diseases , Microglia , Nanostructures , Microglia/drug effects , Microglia/metabolism , Humans , Nanostructures/chemistry , Animals , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism
18.
Brain Behav Immun ; 116: 321-328, 2024 02.
Article in English | MEDLINE | ID: mdl-38157945

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are a subpopulation of innate-like T cells that can be found throughout the body, predominantly in mucosal sites, the lungs and in the peripheral blood. MAIT cells recognize microbial-derived vitamin B (e.g., riboflavin) metabolite antigens that are presented by the major histocompatibility complex class I-like protein, MR1, found on a variety of cell types in the periphery and the CNS. Since their original discovery, MAIT cells have been studied predominantly in their roles in diseases in the periphery; however, it was not until the early 2000s that these cells were first examined for their contributions to disorders of the CNS, with the bulk of the work being done within the past few years. Currently, the MR1/MAIT cell axis has been investigated in only a few neurological diseases including, multiple sclerosis and experimental autoimmune encephalomyelitis, brain cancer/tumors, ischemia, cerebral palsy, general aging and, most recently, Alzheimer's disease. Each of these diseases demonstrates a role for this under-studied innate immune axis in its neuropathology. Together, they highlight the importance of studying the MR1/MAIT cell axis in CNS disorders. Here, we review the contributions of the MR1/MAIT cell axis in the progression or remission of these neurological diseases. This work has shed some light in terms of potentially exploiting the MR1/MAIT cell axis in novel therapeutic applications.


Subject(s)
Central Nervous System Diseases , Mucosal-Associated Invariant T Cells , Humans , Minor Histocompatibility Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Riboflavin/metabolism , Central Nervous System Diseases/metabolism
19.
Biomolecules ; 13(12)2023 11 30.
Article in English | MEDLINE | ID: mdl-38136601

ABSTRACT

Cerebrospinal fluid (CSF) is a biochemical-clinical window into the brain. Unfortunately, its wide dynamic range, low protein concentration, and small sample quantity significantly limit the possibility of using it routinely. Extraventricular drainage (EVD) of CSF allows us to solve quantitative problems and to study the biological role of extracellular vesicles (EVs). In this study, we implemented bioinformatic analysis of our previous data of EVD of CSF and its EVs obtained from congenital hydrocephalus with the aim of identifying a comprehensive list of potential tumor and non-tumor biomarkers of central nervous system diseases. Among all proteins identified, those enriched in EVs are associated with synapses, synaptosomes, and nervous system diseases including gliomas, embryonal tumors, and epilepsy. Among these EV-enriched proteins, given the broad consensus present in the recent scientific literature, we validated syntaxin-binding protein 1 (STXBP1) as a marker of malignancy in EVD of CSF and its EVs from patients with pilocytic astrocytoma and medulloblastoma. Our results show that STXBP1 is negatively enriched in EVs compared to non-tumor diseases and its downregulation correlates with adverse outcomes. Further experiments are needed to validate this and other EV markers in the blood of pediatric patients for translational medicine applications.


Subject(s)
Central Nervous System Diseases , Extracellular Vesicles , Child , Humans , Biomarkers/metabolism , Brain/metabolism , Central Nervous System Diseases/metabolism , Extracellular Vesicles/metabolism , Proteomics/methods
20.
J Control Release ; 364: 46-60, 2023 12.
Article in English | MEDLINE | ID: mdl-37866404

ABSTRACT

The prevalence of central nervous system (CNS) diseases is on the rise as the population ages. The presence of various obstacles, particularly the blood-brain barrier (BBB), poses a challenge for drug delivery to the CNS. An expanding body of study suggests that gut microbiota (GM) plays an important role in CNS diseases. The communication between GM and CNS diseases has received increasing attention. Accumulating evidence indicates that the GM can modulate host signaling pathways to regulate distant organ functions by delivering bioactive substances to host cells via bacterial extracellular vesicles (BEVs). BEVs have emerged as a promising platform for the treatment of CNS diseases due to their nanostructure, ability to penetrate the BBB, as well as their low toxicity, high biocompatibility, ease of modification and large-scale culture. Here, we discuss the biogenesis, internalization mechanism and engineering modification methods of BEVs. We then focus on the use and potential role of BEVs in the treatment of CNS diseases. Finally, we outline the main challenges and future prospects for the application of BEVs in CNS diseases. We hope that the comprehensive understanding of the BEVs-based gut-brain axis will provide new insights into the treatment of CNS diseases.


Subject(s)
Central Nervous System Diseases , Extracellular Vesicles , Humans , Blood-Brain Barrier/metabolism , Central Nervous System Diseases/therapy , Central Nervous System Diseases/metabolism , Extracellular Vesicles/metabolism , Signal Transduction , Biological Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...