Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.834
Filter
1.
Apoptosis ; 29(7-8): 981-1006, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38824478

ABSTRACT

Copper is a trace element required by the organism, but once the level of copper exceeds the threshold, it becomes toxic and even causes death. The underlying mechanisms of copper-induced death are inconclusive, with different studies showing different opinions on the mechanism of copper-induced death. Multiple investigations have shown that copper induces oxidative stress, endoplasmic reticulum stress, nucleolar stress, and proteasome inhibition, all of which can result in cell death. The latest research elucidates a copper-dependent death and denominates it as cuproptosis. Cuproptosis takes place through the combination of copper and lipoylated proteins of the tricarboxylic acid cycle, triggering agglomeration of lipoylated proteins and loss of iron-sulfur cluster proteins, leading to proteotoxic stress and ultimately death. Given the toxicity and necessity of copper, abnormal levels of copper lead to diseases such as neurological diseases and cancer. The development of cancer has a high demand for copper, neurological diseases involve the change of copper contents and the binding of copper to proteins. There is a close relationship between these two kinds of diseases and copper. Here, we summarize the mechanisms of copper-related death, and the association between copper and diseases, to better figure out the influence of copper in cell death and diseases, thus advancing the clinical remedy of these diseases.


Subject(s)
Copper , Neoplasms , Humans , Copper/metabolism , Animals , Neoplasms/metabolism , Neoplasms/pathology , Oxidative Stress , Endoplasmic Reticulum Stress , Cell Death , Apoptosis , Nervous System Diseases/metabolism , Nervous System Diseases/pathology
2.
Curr Opin Neurol ; 37(4): 361-368, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38884636

ABSTRACT

PURPOSE OF REVIEW: The brainstem's complex anatomy and relatively small size means that structural and functional assessment of this structure is done less frequently compared to other brain areas. However, recent years have seen substantial progress in brainstem imaging, enabling more detailed investigations into its structure and function, as well as its role in neuropathology. RECENT FINDINGS: Advancements in ultrahigh field MRI technology have allowed for unprecedented spatial resolution in brainstem imaging, facilitating the new creation of detailed brainstem-specific atlases. Methodological improvements have significantly enhanced the accuracy of physiological (cardiac and respiratory) noise correction within brainstem imaging studies. These technological and methodological advancements have allowed for in-depth analyses of the brainstem's anatomy, including quantitative assessments and examinations of structural connectivity within both gray and white matter. Furthermore, functional studies, including assessments of activation patterns and functional connectivity, have revealed the brainstem's roles in both specialized functions and broader neural integration. Notably, these investigations have identified alterations in brainstem structure and function associated with various neurological disorders. SUMMARY: The aforementioned developments have allowed for a greater appreciation of the importance of the brainstem in the wider context of neuroscience and clinical neurology.


Subject(s)
Brain Stem , Magnetic Resonance Imaging , Humans , Brain Stem/diagnostic imaging , Magnetic Resonance Imaging/methods , Neuroimaging/methods , Nervous System Diseases/diagnostic imaging , Nervous System Diseases/pathology
3.
Cells ; 13(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38891034

ABSTRACT

Astrocytes, the most abundant glial cell type in the brain, play crucial roles in maintaining homeostasis within the central nervous system (CNS). Impairment or abnormalities of typical astrocyte functions in the CNS serve as a causative or contributing factor in numerous neurodevelopmental, neurodegenerative, and neuropsychiatric disorders. Currently, disease-modeling and drug-screening approaches, primarily focused on human astrocytes, rely on human pluripotent stem cell (hPSC)-derived astrocytes. However, it is important to acknowledge that these hPSC-derived astrocytes exhibit notable differences across studies and when compared to their in vivo counterparts. These differences may potentially compromise translational outcomes if not carefully accounted for. This review aims to explore state-of-the-art in vitro models of human astrocyte development, focusing on the developmental processes, functional maturity, and technical aspects of various hPSC-derived astrocyte differentiation protocols. Additionally, it summarizes their successful application in modeling neurological disorders. The discussion extends to recent advancements in the large-scale production of human astrocytes and their application in developing high-throughput assays conducive to therapeutic drug discovery.


Subject(s)
Astrocytes , Cell Differentiation , Nervous System Diseases , Pluripotent Stem Cells , Humans , Astrocytes/metabolism , Astrocytes/cytology , Nervous System Diseases/pathology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Translational Research, Biomedical , Animals
4.
Int J Biochem Cell Biol ; 173: 106614, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38944234

ABSTRACT

The human body is commonly exposed to bisphenol A (BPA), which is widely used in consumer and industrial products. BPA is an endocrine-disrupting chemical that has adverse effects on human health. In particular, many studies have shown that BPA can cause various neurological disorders by affecting brain development and neural function during prenatal, infancy, childhood, and adulthood exposure. In this review, we discussed the correlation between BPA and neurological disorders based on molecular cell biology, neurophysiology, and behavioral studies of the effects of BPA on brain development and function. Recent studies, both animal and epidemiological, strongly indicate that BPA significantly impacts brain development and function. It hinders neural processes, such as proliferation, migration, and differentiation during development, affecting synaptic formation and activity. As a result, BPA is implicated in neurodevelopmental and neuropsychiatric disorders like autism spectrum disorder (ASD), attention-deficit hyperactivity disorder (ADHD), and schizophrenia.


Subject(s)
Benzhydryl Compounds , Nervous System Diseases , Phenols , Humans , Benzhydryl Compounds/adverse effects , Benzhydryl Compounds/toxicity , Phenols/toxicity , Phenols/adverse effects , Animals , Nervous System Diseases/chemically induced , Nervous System Diseases/pathology , Endocrine Disruptors/adverse effects , Endocrine Disruptors/toxicity , Brain/drug effects , Brain/growth & development , Brain/pathology
5.
Cell Genom ; 4(7): 100585, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38942022

ABSTRACT

Sexual dimorphism, differences between males and females of the same species, is widespread in mammals. However, good animal models to study human sexually dimorphic phenotypes are currently lacking. In this issue, DeCasien et al.1 explore the potential of rhesus macaque as a model for investigating sexually dimorphic traits in the human brain.


Subject(s)
Disease Models, Animal , Macaca mulatta , Nervous System Diseases , Sex Characteristics , Animals , Nervous System Diseases/pathology , Nervous System Diseases/physiopathology , Humans , Female , Male
6.
J Vet Sci ; 25(4): e45, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38910307

ABSTRACT

IMPORTANCE: Although the role of bovine coronavirus (BCoV) in calf diarrhea and respiratory disorders is well documented, its contribution to neurological diseases is unclear. OBJECTIVE: This study conducted virological investigations of calves showing diarrhea and respiratory and neurological signs. METHODS: An outbreak of diarrhea, respiratory, and neurological disorders occurred among the 12 calves in July 2022 in Istanbul, Türkiye. Two of these calves exhibited neurological signs and died a few days after the appearance of symptoms. One of these calves was necropsied and analyzed using molecular and histopathological tests. RESULTS: BCoV RNA was detected in the brain, lung, spleen, liver, and intestine of the calf that had neurological signs by real-time reverse transcription polymerase chain reaction. Immunostaining was also observed in the intestine and brain. A 622 bp S1 gene product was noted on gel electrophoresis only in the brain. Phylogenetic analysis indicated that the BCoV detected in this study had a high proximity to the BCoV strain GIb with 99.19% nucleotide sequence homology to the strains detected in Poland, Israel, Türkiye, and France. No distinct genetic lineages were observed when the brain isolate was compared with the respiratory and enteric strains reported to GenBank. In addition, the highest identity (98,72%) was obtained with the HECV 4408 and L07748 strains of human coronaviruses. CONCLUSIONS AND RELEVANCE: The strain detected in a calf brain belongs to the GIb-European lineage and shares high sequence homology with BCoV strains detected in Europe and Israel. In addition, the similarity between the human coronaviruses (4408 and L07748) raises questions about the zoonotic potential of the strains detected in this study.


Subject(s)
Brain , Cattle Diseases , Coronavirus Infections , Coronavirus, Bovine , Phylogeny , Animals , Cattle , Coronavirus, Bovine/genetics , Coronavirus, Bovine/isolation & purification , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/pathology , Cattle Diseases/virology , Cattle Diseases/pathology , Brain/virology , Brain/pathology , Turkey/epidemiology , Disease Outbreaks/veterinary , Pneumonia, Viral/veterinary , Pneumonia, Viral/virology , Pneumonia, Viral/pathology , Nervous System Diseases/veterinary , Nervous System Diseases/virology , Nervous System Diseases/pathology
7.
Adv Drug Deliv Rev ; 210: 115344, 2024 07.
Article in English | MEDLINE | ID: mdl-38810702

ABSTRACT

Brain organoids hold great potential for modeling human brain development and pathogenesis. They recapitulate certain aspects of the transcriptional trajectory, cellular diversity, tissue architecture and functions of the developing brain. In this review, we explore the engineering strategies to control the molecular-, cellular- and tissue-level inputs to achieve high-fidelity brain organoids. We review the application of brain organoids in neural disorder modeling and emerging bioengineering methods to improve data collection and feature extraction at multiscale. The integration of multiscale engineering strategies and analytical methods has significant potential to advance insight into neurological disorders and accelerate drug development.


Subject(s)
Brain , Organoids , Humans , Brain/metabolism , Brain/cytology , Animals , Models, Biological , Nervous System Diseases/pathology , Tissue Engineering/methods , Bioengineering/methods
8.
Cell Death Dis ; 15(4): 269, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627369

ABSTRACT

Most of the patients affected by neuronopathic forms of Mucopolysaccharidosis type II (MPS II), a rare lysosomal storage disorder caused by defects in iduronate-2-sulfatase (IDS) activity, exhibit early neurological defects associated with white matter lesions and progressive behavioural abnormalities. While neuronal degeneration has been largely described in experimental models and human patients, more subtle neuronal pathogenic defects remain still underexplored. In this work, we discovered that the axon guidance receptor Deleted in Colorectal Cancer (Dcc) is significantly dysregulated in the brain of ids mutant zebrafish since embryonic stages. In addition, thanks to the establishment of neuronal-enriched primary cell cultures, we identified defective proteasomal degradation as one of the main pathways underlying Dcc upregulation in ids mutant conditions. Furthermore, ids mutant fish-derived primary neurons displayed higher levels of polyubiquitinated proteins and P62, suggesting a wider defect in protein degradation. Finally, we show that ids mutant larvae display an atypical response to anxiety-inducing stimuli, hence mimicking one of the characteristic features of MPS II patients. Our study provides an additional relevant frame to MPS II pathogenesis, supporting the concept that multiple developmental defects concur with early childhood behavioural abnormalities.


Subject(s)
Iduronate Sulfatase , Mucopolysaccharidosis II , Nervous System Diseases , Animals , Axon Guidance , Brain/metabolism , Iduronate Sulfatase/metabolism , Mucopolysaccharidosis II/metabolism , Nervous System Diseases/pathology , Zebrafish/metabolism
9.
Curr Opin Microbiol ; 79: 102474, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615394

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019, contributes to neurological pathologies in nearly 30% of patients, extending beyond respiratory symptoms. These manifestations encompass disorders of both the peripheral and central nervous systems, causing among others cerebrovascular issues and psychiatric manifestations during the acute and/or post-acute infection phases. Despite ongoing research, uncertainties persist about the precise mechanism the virus uses to infiltrate the central nervous system and the involved entry portals. This review discusses the potential entry routes, including hematogenous and anterograde transport. Furthermore, we explore variations in neurotropism, neurovirulence, and neurological manifestations among pandemic-associated variants of concern. In conclusion, SARS-CoV-2 can infect numerous cells within the peripheral and central nervous system, provoke inflammatory responses, and induce neuropathological changes.


Subject(s)
COVID-19 , SARS-CoV-2 , Viral Tropism , Humans , COVID-19/virology , COVID-19/pathology , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , Central Nervous System/virology , Central Nervous System/pathology , Animals , Nervous System Diseases/virology , Nervous System Diseases/pathology , Nervous System Diseases/physiopathology , Virus Internalization
10.
Expert Rev Mol Med ; 26: e11, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38682637

ABSTRACT

Long non-coding RNAs (lncRNAs) are progressively being perceived as prominent molecular agents controlling multiple aspects of neuronal (patho)physiology. Amongst these is the HOX transcript antisense intergenic RNA, often abbreviated as HOTAIR. HOTAIR epigenetically regulates its target genes via its interaction with two different chromatin-modifying agents; histone methyltransferase polycomb-repressive complex 2 and histone demethylase lysine-specific demethylase 1. Parenthetically, HOTAIR elicits trans-acting sponging function against multiple micro-RNA species. Oncological research studies have confirmed the pathogenic functions of HOTAIR in multiple cancer types, such as gliomas and proposed it as a pro-oncological lncRNA. In fact, its expression has been suggested to be a predictor of the severity/grade of gliomas, and as a prognostic biomarker. Moreover, a propound influence of HOTAIR in other aspects of brain heath and disease states is just beginning to be unravelled. The objective of this review is to recapitulate all the relevant data pertaining to the regulatory roles of HOTAIR in neuronal (patho)physiology. To this end, we discuss the pathogenic mechanisms of HOTAIR in multiple neuronal diseases, such as neurodegeneration, traumatic brain injury and neuropsychiatric disorders. Finally, we also summarize the results from the studies incriminating HOTAIR in the pathogeneses of gliomas and other brain cancers. Implications of HOTAIR serving as a suitable therapeutic target in neuropathologies are also discussed.


Subject(s)
RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , Animals , Prognosis , Epigenesis, Genetic , Biomarkers , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Nervous System Diseases/therapy , Nervous System Diseases/pathology , Glioma/genetics , Glioma/pathology , Glioma/therapy , Glioma/metabolism
11.
Cells ; 13(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38667285

ABSTRACT

Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), stroke, and aneurysms, are characterized by the abnormal accumulation and aggregation of disease-causing proteins in the brain and spinal cord. Recent research suggests that proteins linked to these conditions can be secreted and transferred among cells using exosomes. The transmission of abnormal protein buildup and the gradual degeneration in the brains of impacted individuals might be supported by these exosomes. Furthermore, it has been reported that neuroprotective functions can also be attributed to exosomes in neurodegenerative diseases. The potential neuroprotective functions may play a role in preventing the formation of aggregates and abnormal accumulation of proteins associated with the disease. The present review summarizes the roles of exosomes in neurodegenerative diseases as well as elucidating their therapeutic potential in AD, PD, ALS, HD, stroke, and aneurysms. By elucidating these two aspects of exosomes, valuable insights into potential therapeutic targets for treating neurodegenerative diseases may be provided.


Subject(s)
Exosomes , Exosomes/metabolism , Humans , Animals , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Vascular Diseases/metabolism , Vascular Diseases/pathology , Nervous System Diseases/metabolism , Nervous System Diseases/pathology
12.
Front Biosci (Landmark Ed) ; 29(4): 142, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38682185

ABSTRACT

Innate lymphocytes, including microglial cells, astrocytes, and oligodendrocytes, play a crucial role in initiating neuroinflammatory reactions inside the central nervous system (CNS). The prime focus of this paper is on the involvement and interplay of neurons and glial cells in neurological disorders such as Alzheimer's Disease (AD), Autism Spectrum Disorder (ASD), epilepsy, and multiple sclerosis (MS). In this review, we explore the specific contributions of microglia and astrocytes and analyzes multiple pathways implicated in neuroinflammation and disturbances in excitatory and inhibitory processes. Firstly, we elucidate the mechanisms through which toxic protein accumulation in AD results in synaptic dysfunction and deregulation of the immune system and examines the roles of microglia, astrocytes, and hereditary factors in the pathogenesis of the disease. Secondly, we focus on ASD and the involvement of glial cells in the development of the nervous system and the formation of connections between neurons and investigates the genetic connections associated with these processes. Lastly, we also address the participation of glial cells in epilepsy and MS, providing insights into their pivotal functions in both conditions. We also tried to give an overview of seven different pathways like toll-like receptor signalling pathway, MyD88-dependent and independent pathway, etc and its relevance in the context with these neurological disorders. In this review, we also explore the role of activated glial cells in AD, ASD, epilepsy, and MS which lead to neuroinflammation. Even we focus on excitatory and inhibitory imbalance in all four neurological disorders as imbalance affect the proper functioning of neuronal circuits. Finally, this review concludes that there is necessity for additional investigation on glial cells and their involvement in neurological illnesses.


Subject(s)
Nervous System Diseases , Neuroglia , Neurons , Animals , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Astrocytes/metabolism , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/genetics , Cell Communication , Epilepsy/genetics , Epilepsy/metabolism , Epilepsy/physiopathology , Microglia/metabolism , Multiple Sclerosis/metabolism , Multiple Sclerosis/genetics , Multiple Sclerosis/physiopathology , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Neuroglia/metabolism , Neuroinflammatory Diseases/metabolism , Neurons/metabolism , Signal Transduction
13.
Medicine (Baltimore) ; 103(14): e37721, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38579030

ABSTRACT

RATIONALE: White matter lesions (WMLs) are structural changes in the brain that manifest as demyelination in the central nervous system pathologically. Vasogenic WMLs are the most prevalent type, primarily associated with advanced age and cerebrovascular risk factors. Conversely, immunogenic WMLs, typified by multiple sclerosis (MS), are more frequently observed in younger patients. It is crucial to distinguish between these 2 etiologies. Furthermore, in cases where multiple individuals exhibit WMLs within 1 family, genetic testing may offer a significant diagnostic perspective. PATIENT CONCERNS: A 25-year-old male presented to the Department of Neurology with recurrent headaches. He was healthy previously and the neurological examination was negative. Brain magnetic resonance imaging (MRI) showed widespread white matter hyperintensity lesions surrounding the ventricles and subcortical regions on T2-weighted and T2 fluid-attenuated inversion recovery images, mimicking immunogenic disease-MS. DIAGNOSES: The patient was diagnosed with a patent foramen ovale, which could explain his headache syndrome. Genetic testing unveiled a previously unidentified missense mutation in the SERPINC1 gene in the patient and his father. The specific abnormal laboratory finding was a reduction in antithrombin III activity, and the decrease may serve as the underlying cause for the presence of multiple intracranial WMLs observed in both the patient and his father. INTERVENTIONS: The patient received percutaneous patent foramen ovale closure surgery and took antiplatelet drug recommended by cardiologists and was followed up for 1 month and 6 months after operation. OUTCOMES: While the lesions on MRI remain unchanging during follow-up, the patient reported a significant relief in headaches compared to the initial presentation. LESSONS: This case introduces a novel perspective on the etiology of cerebral WMLs, suggesting that hereditary antithrombin deficiency (ATD) could contribute to altered blood composition and may serve as an underlying cause in certain individuals with asymptomatic WMLs.


Subject(s)
Antithrombin III Deficiency , Foramen Ovale, Patent , Multiple Sclerosis , Nervous System Diseases , Vascular Diseases , White Matter , Male , Humans , Adult , White Matter/diagnostic imaging , White Matter/pathology , Foramen Ovale, Patent/pathology , Antithrombin III/genetics , Antithrombin III Deficiency/complications , Antithrombin III Deficiency/genetics , Antithrombin III Deficiency/pathology , Brain/pathology , Magnetic Resonance Imaging/methods , Vascular Diseases/pathology , Nervous System Diseases/pathology , Multiple Sclerosis/diagnosis , Headache , Mutation , Antithrombins
14.
J Vet Intern Med ; 38(3): 1808-1814, 2024.
Article in English | MEDLINE | ID: mdl-38669583

ABSTRACT

BACKGROUND: In 2020, a novel neurologic disease was observed in juvenile Quarter Horses (QHs) in North America. It was unknown if this was an aberrant manifestation of another previously described neurological disorder in foals, such as equine neuroaxonal dystrophy/equine degenerative myeloencephalopathy (eNAD/EDM). HYPOTHESIS/OBJECTIVES: To describe the clinical findings, outcomes, and postmortem changes with Equine Juvenile Spinocerebellar Ataxia (EJSCA), differentiate the disease from other similar neurological disorders, and determine a mode of inheritance. ANIMALS: Twelve neurologically affected QH foals and the dams. METHODS: Genomic DNA was isolated and pedigrees were manually constructed. RESULTS: All foals (n = 12/12) had a history of acute onset of neurological deficits with no history of trauma. Neurological deficits were characterized by asymmetrical spinal ataxia, with pelvic limbs more severely affected than thoracic limbs. Clinicopathological abnormalities included high serum activity of gamma-glutamyl transferase and hyperglycemia. All foals became recumbent (median, 3 days: [0-18 days]), which necessitated humane euthanasia (n = 11/12, 92%; the remaining case was found dead). Histological evaluation at postmortem revealed dilated myelin sheaths and digestion chambers within the spinal cord, most prominently in the dorsal spinocerebellar tracts. Pedigree analysis revealed a likely autosomal recessive mode of inheritance. CONCLUSIONS AND CLINICAL IMPORTANCE: EJSCA is a uniformly fatal, rapidly progressive, likely autosomal recessive neurological disease of QHs <1 month of age in North America that is etiologically distinct from other clinically similar neurological disorders. Once the causative variant for EJSCA is validated, carriers can be identified through genetic testing to inform breeding decisions.


Subject(s)
Horse Diseases , Pedigree , Animals , Horses , Horse Diseases/genetics , Horse Diseases/pathology , Male , Female , North America , Spinocerebellar Ataxias/veterinary , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology , Nervous System Diseases/veterinary , Nervous System Diseases/genetics , Nervous System Diseases/pathology
15.
Signal Transduct Target Ther ; 9(1): 112, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670977

ABSTRACT

The induced pluripotent stem cell (iPSC) technology has transformed in vitro research and holds great promise to advance regenerative medicine. iPSCs have the capacity for an almost unlimited expansion, are amenable to genetic engineering, and can be differentiated into most somatic cell types. iPSCs have been widely applied to model human development and diseases, perform drug screening, and develop cell therapies. In this review, we outline key developments in the iPSC field and highlight the immense versatility of the iPSC technology for in vitro modeling and therapeutic applications. We begin by discussing the pivotal discoveries that revealed the potential of a somatic cell nucleus for reprogramming and led to successful generation of iPSCs. We consider the molecular mechanisms and dynamics of somatic cell reprogramming as well as the numerous methods available to induce pluripotency. Subsequently, we discuss various iPSC-based cellular models, from mono-cultures of a single cell type to complex three-dimensional organoids, and how these models can be applied to elucidate the mechanisms of human development and diseases. We use examples of neurological disorders, coronavirus disease 2019 (COVID-19), and cancer to highlight the diversity of disease-specific phenotypes that can be modeled using iPSC-derived cells. We also consider how iPSC-derived cellular models can be used in high-throughput drug screening and drug toxicity studies. Finally, we discuss the process of developing autologous and allogeneic iPSC-based cell therapies and their potential to alleviate human diseases.


Subject(s)
COVID-19 , Cellular Reprogramming , Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/cytology , Cellular Reprogramming/genetics , SARS-CoV-2/genetics , Cell Differentiation/genetics , Neoplasms/genetics , Neoplasms/therapy , Neoplasms/pathology , Regenerative Medicine , Nervous System Diseases/therapy , Nervous System Diseases/genetics , Nervous System Diseases/pathology
16.
17.
Pharmacol Res ; 203: 107149, 2024 May.
Article in English | MEDLINE | ID: mdl-38518830

ABSTRACT

Neuronal health is closely linked to the homeostasis of intracellular organelles, and organelle dysfunction affects the pathological progression of neurological diseases. In contrast to isolated cellular compartments, a growing number of studies have found that organelles are largely interdependent structures capable of communicating through membrane contact sites (MCSs). MCSs have been identified as key pathways mediating inter-organelle communication crosstalk in neurons, and their alterations have been linked to neurological disease pathology. The endoplasmic reticulum (ER) is a membrane-bound organelle capable of forming an extensive network of pools and tubules with important physiological functions within neurons. There are multiple MCSs between the ER and other organelles and the plasma membrane (PM), which regulate a variety of cellular processes. In this review, we focus on ER-organelle MCSs and their role in a variety of neurological diseases. We compared the biological effects between different tethering proteins and the effects of their respective disease counterparts. We also discuss how altered ER-organelle contacts may affect disease pathogenesis. Therefore, understanding the molecular mechanisms of ER-organelle MCSs in neuronal homeostasis will lay the foundation for the development of new therapies targeting ER-organelle contacts.


Subject(s)
Endoplasmic Reticulum , Nervous System Diseases , Signal Transduction , Humans , Endoplasmic Reticulum/metabolism , Animals , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Neurons/metabolism , Organelles/metabolism
18.
Rev Neurosci ; 35(5): 489-502, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-38440811

ABSTRACT

Since Cajal introduced dendritic spines in the 19th century, they have attained considerable attention, especially in neuropsychiatric and neurologic disorders. Multiple roles of dendritic spine malfunction and pathology in the progression of various diseases have been reported. Thus, it is inevitable to consider these structures as new therapeutic targets for treating neuropsychiatric and neurologic disorders such as autism spectrum disorders, schizophrenia, dementia, Down syndrome, etc. Therefore, we attempted to prepare a narrative review of the literature regarding the role of dendritic spines in the pathogenesis of aforementioned diseases and to shed new light on their pathophysiology.


Subject(s)
Dendritic Spines , Nervous System Diseases , Neurodevelopmental Disorders , Humans , Dendritic Spines/pathology , Nervous System Diseases/physiopathology , Nervous System Diseases/pathology , Nervous System Diseases/etiology , Animals , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/physiopathology
19.
Semin Nucl Med ; 54(2): 237-246, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38365546

ABSTRACT

Sex differences in brain physiology and the mechanisms of drug action have been extensively reported. These biological variances, from structure to hormonal and genetic aspects, can profoundly influence healthy functioning and disease mechanisms and might have implications for treatment and drug development. Molecular neuroimaging techniques may help to disclose sex's impact on brain functioning, as well as the neuropathological changes underpinning several diseases. This narrative review summarizes recent lines of evidence based on PET and SPECT imaging, highlighting sex differences in normal conditions and various neurological disorders.


Subject(s)
Nervous System Diseases , Neuroimaging , Female , Humans , Male , Neuroimaging/methods , Brain/diagnostic imaging , Nervous System Diseases/diagnostic imaging , Nervous System Diseases/pathology , Women's Health , Tomography, Emission-Computed, Single-Photon
20.
Nat Commun ; 15(1): 1667, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38396116

ABSTRACT

Microglia nodules (HLA-DR+ cell clusters) are associated with brain pathology. In this post-mortem study, we investigated whether they represent the first stage of multiple sclerosis (MS) lesion formation. We show that microglia nodules are associated with more severe MS pathology. Compared to microglia nodules in stroke, those in MS show enhanced expression of genes previously found upregulated in MS lesions. Furthermore, genes associated with lipid metabolism, presence of T and B cells, production of immunoglobulins and cytokines, activation of the complement cascade, and metabolic stress are upregulated in microglia nodules in MS. Compared to stroke, they more frequently phagocytose oxidized phospholipids and possess a more tubular mitochondrial network. Strikingly, in MS, some microglia nodules encapsulate partially demyelinated axons. Taken together, we propose that activation of microglia nodules in MS by cytokines and immunoglobulins, together with phagocytosis of oxidized phospholipids, may lead to a microglia phenotype prone to MS lesion formation.


Subject(s)
Multiple Sclerosis , Nervous System Diseases , Stroke , Humans , Multiple Sclerosis/pathology , Microglia/metabolism , Nervous System Diseases/pathology , Stroke/pathology , Cytokines/metabolism , Immunoglobulins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL