Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28.981
Filter
1.
BMC Med ; 22(1): 266, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38951846

ABSTRACT

BACKGROUND: Benzodiazepine use is common, particularly in older adults. Benzodiazepines have well-established acute adverse effects on cognition, but long-term effects on neurodegeneration and dementia risk remain uncertain. METHODS: We included 5443 cognitively healthy (MMSE ≥ 26) participants from the population-based Rotterdam Study (57.4% women, mean age 70.6 years). Benzodiazepine use from 1991 until baseline (2005-2008) was derived from pharmacy dispensing records, from which we determined drug type and cumulative dose. Benzodiazepine use was defined as prescription of anxiolytics (ATC-code: N05BA) or sedative-hypnotics (ATC-code: N05CD) between inception of pharmacy records and study baseline. Cumulative dose was calculated as the sum of the defined daily doses for all prescriptions. We determined the association with dementia risk until 2020 using Cox regression. Among 4836 participants with repeated brain MRI, we further determined the association of benzodiazepine use with changes in neuroimaging markers using linear mixed models. RESULTS: Of all 5443 participants, 2697 (49.5%) had used benzodiazepines at any time in the 15 years preceding baseline, of whom 1263 (46.8%) used anxiolytics, 530 (19.7%) sedative-hypnotics, and 904 (33.5%) used both; 345 (12.8%) participants were still using at baseline assessment. During a mean follow-up of 11.2 years, 726 participants (13.3%) developed dementia. Overall, use of benzodiazepines was not associated with dementia risk compared to never use (HR [95% CI]: 1.06 [0.90-1.25]), irrespective of cumulative dose. Risk estimates were somewhat higher for any use of anxiolytics than for sedative-hypnotics (HR 1.17 [0.96-1.41] vs 0.92 [0.70-1.21]), with strongest associations for high cumulative dose of anxiolytics (HR [95% CI] 1.33 [1.04-1.71]). In imaging analyses, current use of benzodiazepine was associated cross-sectionally with lower brain volumes of the hippocampus, amygdala, and thalamus and longitudinally with accelerated volume loss of the hippocampus and to a lesser extent amygdala. However, imaging findings did not differ by type of benzodiazepines or cumulative dose. CONCLUSIONS: In this population-based sample of cognitively healthy adults, overall use of benzodiazepines was not associated with increased dementia risk, but potential class-dependent adverse effects and associations with subclinical markers of neurodegeneration may warrant further investigation.


Subject(s)
Benzodiazepines , Dementia , Humans , Female , Dementia/epidemiology , Dementia/chemically induced , Male , Aged , Benzodiazepines/adverse effects , Benzodiazepines/administration & dosage , Middle Aged , Magnetic Resonance Imaging , Netherlands/epidemiology , Aged, 80 and over , Neuroimaging , Brain/diagnostic imaging , Brain/drug effects , Brain/pathology , Prospective Studies , Neurodegenerative Diseases/epidemiology , Neurodegenerative Diseases/chemically induced , Hypnotics and Sedatives/adverse effects , Risk Factors
2.
Adv Protein Chem Struct Biol ; 141: 123-176, 2024.
Article in English | MEDLINE | ID: mdl-38960472

ABSTRACT

Metalloproteins represents more than one third of human proteome, with huge variation in physiological functions and pathological implications, depending on the metal/metals involved and tissue context. Their functions range from catalysis, bioenergetics, redox, to DNA repair, cell proliferation, signaling, transport of vital elements, and immunity. The human metalloproteomic studies revealed that many families of metalloproteins along with individual metalloproteins are dysregulated under several clinical conditions. Also, several sorts of interaction between redox- active or redox- inert metalloproteins are observed in health and disease. Metalloproteins profiling shows distinct alterations in neurodegenerative diseases, cancer, inflammation, infection, diabetes mellitus, among other diseases. This makes metalloproteins -either individually or as families- a promising target for several therapeutic approaches. Inhibitors and activators of metalloenzymes, metal chelators, along with artificial metalloproteins could be versatile in diagnosis and treatment of several diseases, in addition to other biomedical and industrial applications.


Subject(s)
Metalloproteins , Proteomics , Humans , Metalloproteins/metabolism , Neoplasms/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology
3.
Clin Exp Med ; 24(1): 142, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38958690

ABSTRACT

This comprehensive exploration delves into the pivotal role of microRNAs (miRNAs) within the intricate tapestry of cellular regulation. As potent orchestrators of gene expression, miRNAs exhibit diverse functions in cellular processes, extending their influence from the nucleus to the cytoplasm. The complex journey of miRNA biogenesis, involving transcription, processing, and integration into the RNA-induced silencing complex, showcases their versatility. In the cytoplasm, mature miRNAs finely tune cellular functions by modulating target mRNA expression, while their reach extends into the nucleus, influencing transcriptional regulation and epigenetic modifications. Dysregulation of miRNAs becomes apparent in various pathologies, such as cancer, autoimmune diseases, and inflammatory conditions. The adaptability of miRNAs to environmental signals, interactions with transcription factors, and involvement in intricate regulatory networks underscore their significance. DNA methylation and histone modifications adds depth to understanding the dynamic regulation of miRNAs. Mechanisms like competition with RNA-binding proteins, sponging, and the control of miRNA levels through degradation and editing contribute to this complex regulation process. In this review, we mainly focus on how dysregulation of miRNA expression can be related with skin-related autoimmune and autoinflammatory diseases, arthritis, cardiovascular diseases, inflammatory bowel disease, autoimmune and autoinflammatory diseases, and neurodegenerative disorders. We also emphasize the multifaceted roles of miRNAs, urging continued research to unravel their complexities. The mechanisms governing miRNA functions promise advancements in therapeutic interventions and enhanced insights into cellular dynamics in health and disease.


Subject(s)
Gene Expression Regulation , Inflammation , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Inflammation/genetics , Autoimmune Diseases/genetics , Epigenesis, Genetic , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism
4.
Acta Neuropathol ; 148(1): 2, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38980441

ABSTRACT

Proteolytic cell surface release ('shedding') of the prion protein (PrP), a broadly expressed GPI-anchored glycoprotein, by the metalloprotease ADAM10 impacts on neurodegenerative and other diseases in animal and in vitro models. Recent studies employing the latter also suggest shed PrP (sPrP) to be a ligand in intercellular communication and critically involved in PrP-associated physiological tasks. Although expectedly an evolutionary conserved event, and while soluble forms of PrP are present in human tissues and body fluids, for the human body neither proteolytic PrP shedding and its cleavage site nor involvement of ADAM10 or the biological relevance of this process have been demonstrated thus far. In this study, cleavage site prediction and generation (plus detailed characterization) of sPrP-specific antibodies enabled us to identify PrP cleaved at tyrosin 226 as the physiological and apparently strictly ADAM10-dependent shed form in humans. Using cell lines, neural stem cells and brain organoids, we show that shedding of human PrP can be stimulated by PrP-binding ligands without targeting the protease, which may open novel therapeutic perspectives. Site-specific antibodies directed against human sPrP also detect the shed form in brains of cattle, sheep and deer, hence in all most relevant species naturally affected by fatal and transmissible prion diseases. In human and animal prion diseases, but also in patients with Alzheimer`s disease, sPrP relocalizes from a physiological diffuse tissue pattern to intimately associate with extracellular aggregated deposits of misfolded proteins characteristic for the respective pathological condition. Findings and research tools presented here will accelerate novel insight into the roles of PrP shedding (as a process) and sPrP (as a released factor) in neurodegeneration and beyond.


Subject(s)
ADAM10 Protein , Amyloid Precursor Protein Secretases , Neurodegenerative Diseases , Humans , ADAM10 Protein/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Amyloid Precursor Protein Secretases/metabolism , Animals , Prion Proteins/metabolism , Membrane Proteins/metabolism , Brain/metabolism , Brain/pathology , Antibodies
5.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 40: e20240008, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38952174

ABSTRACT

The numerous and varied forms of neurodegenerative illnesses provide a considerable challenge to contemporary healthcare. The emergence of artificial intelligence has fundamentally changed the diagnostic picture by providing effective and early means of identifying these crippling illnesses. As a subset of computational intelligence, machine-learning algorithms have become very effective tools for the analysis of large datasets that include genetic, imaging, and clinical data. Moreover, multi-modal data integration, which includes information from brain imaging (MRI, PET scans), genetic profiles, and clinical evaluations, is made easier by computational intelligence. A thorough knowledge of the course of the illness is made possible by this consolidative method, which also facilitates the creation of predictive models for early medical evaluation and outcome prediction. Furthermore, there has been a great deal of promise shown by the use of artificial intelligence to neuroimaging analysis. Sophisticated image processing methods combined with machine learning algorithms make it possible to identify functional and structural anomalies in the brain, which often act as early indicators of neurodegenerative diseases. This chapter examines how computational intelligence plays a critical role in improving the diagnosis of neurodegenerative diseases such as Parkinson's, Alzheimer's, etc. To sum up, computational intelligence provides a revolutionary approach for improving the identification of neurodegenerative illnesses. In the battle against these difficult disorders, embracing and improving these computational techniques will surely pave the path for more individualized therapy and more therapies that are successful.


Subject(s)
Computational Biology , Machine Learning , Neurodegenerative Diseases , Neuroimaging , Humans , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/diagnostic imaging , Computational Biology/methods , Neuroimaging/methods , Algorithms , Artificial Intelligence , Brain/diagnostic imaging , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods
7.
J Cell Biochem ; 125(7): e30624, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38946063

ABSTRACT

The 14-3-3 family of proteins are highly conserved acidic eukaryotic proteins (25-32 kDa) abundantly present in the body. Through numerous binding partners, the 14-3-3 is responsible for many essential cellular pathways, such as cell cycle regulation and gene transcription control. Hence, its dysregulation has been linked to the onset of critical illnesses such as cancers, neurodegenerative diseases and viral infections. Interestingly, explorative studies have revealed an inverse correlation of 14-3-3 protein in cancer and neurodegenerative diseases, and the direct manipulation of 14-3-3 by virus to enhance infection capacity has dramatically extended its significance. Of these, COVID-19 has been linked to the 14-3-3 proteins by the interference of the SARS-CoV-2 nucleocapsid (N) protein during virion assembly. Given its predisposition towards multiple essential host signalling pathways, it is vital to understand the holistic interactions between the 14-3-3 protein to unravel its potential therapeutic unit in the future. As such, the general structure and properties of the 14-3-3 family of proteins, as well as their known biological functions and implications in cancer, neurodegeneration, and viruses, were covered in this review. Furthermore, the potential therapeutic target of 14-3-3 proteins in the associated diseases was discussed.


Subject(s)
14-3-3 Proteins , COVID-19 , Neoplasms , Neurodegenerative Diseases , 14-3-3 Proteins/metabolism , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/virology , Neoplasms/metabolism , Neoplasms/virology , Neoplasms/genetics , COVID-19/metabolism , COVID-19/virology , SARS-CoV-2/metabolism , Virus Diseases/metabolism , Virus Diseases/virology , Virus Diseases/genetics
8.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39000036

ABSTRACT

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


Subject(s)
Air Pollutants , Air Pollution , Alzheimer Disease , Humans , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Air Pollution/adverse effects , Air Pollutants/adverse effects , Air Pollutants/toxicity , Risk Factors , Animals , Particulate Matter/adverse effects , Oxidative Stress , Neurodegenerative Diseases/etiology , Environmental Exposure/adverse effects , Brain/pathology , Brain/metabolism
9.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39000116

ABSTRACT

Metabolic endotoxemia is a severe health problem for residents in developed countries who follow a Western diet, disrupting intestinal microbiota and the whole organism's homeostasis. Although the effect of endotoxin on the human immune system is well known, its long-term impact on the human body, lasting many months or even years, is unknown. This is due to the difficulty of conducting in vitro and in vivo studies on the prolonged effect of endotoxin on the central nervous system. In this article, based on the available literature, we traced the path of endotoxin from the intestines to the blood through the intestinal epithelium and factors promoting the development of metabolic endotoxemia. The presence of endotoxin in the bloodstream and the inflammation it induces may contribute to lowering the blood-brain barrier, potentially allowing its penetration into the central nervous system; although, the theory is still controversial. Microglia, guarding the central nervous system, are the first line of defense and respond to endotoxin with activation, which may contribute to the development of neurodegenerative diseases. We traced the pro-inflammatory role of endotoxin in neurodegenerative diseases and its impact on the epigenetic regulation of microglial phenotypes.


Subject(s)
Endotoxemia , Endotoxins , Gastrointestinal Microbiome , Neurodegenerative Diseases , Endotoxemia/metabolism , Endotoxemia/etiology , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/etiology , Animals , Endotoxins/metabolism , Microglia/metabolism , Microglia/pathology , Blood-Brain Barrier/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/microbiology , Inflammation/metabolism
10.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000122

ABSTRACT

Among the various drug discovery methods, a very promising modern approach consists in designing multi-target-directed ligands (MTDLs) able to modulate multiple targets of interest, including the pathways where hydrogen sulfide (H2S) is involved. By incorporating an H2S donor moiety into a native drug, researchers have been able to simultaneously target multiple therapeutic pathways, resulting in improved treatment outcomes. This review gives the reader some pills of successful multi-target H2S-donating molecules as worthwhile tools to combat the multifactorial nature of complex disorders, such as inflammatory-based diseases and cancer, as well as cardiovascular, metabolic, and neurodegenerative disorders.


Subject(s)
Hydrogen Sulfide , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Humans , Animals , Ligands , Drug Discovery/methods , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism
11.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000142

ABSTRACT

Overexpression of protein tyrosine phosphatase 1B (PTP1B) disrupts signaling pathways and results in numerous human diseases. In particular, its involvement has been well documented in the pathogenesis of metabolic disorders (diabetes mellitus type I and type II, fatty liver disease, and obesity); neurodegenerative diseases (Alzheimer's disease, Parkinson's disease); major depressive disorder; calcific aortic valve disease; as well as several cancer types. Given this multitude of therapeutic applications, shortly after identification of PTP1B and its role, the pursuit to introduce safe and selective enzyme inhibitors began. Regrettably, efforts undertaken so far have proved unsuccessful, since all proposed PTP1B inhibitors failed, or are yet to complete, clinical trials. Intending to aid introduction of the new generation of PTP1B inhibitors, this work collects and organizes the current state of the art. In particular, this review intends to elucidate intricate relations between numerous diseases associated with the overexpression of PTP1B, as we believe that it is of the utmost significance to establish and follow a brand-new holistic approach in the treatment of interconnected conditions. With this in mind, this comprehensive review aims to validate the PTP1B enzyme as a promising molecular target, and to reinforce future research in this direction.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Humans , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Neoplasms/metabolism , Neoplasms/enzymology , Neoplasms/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/enzymology , Enzyme Inhibitors/therapeutic use , Enzyme Inhibitors/pharmacology , Metabolic Diseases/metabolism , Metabolic Diseases/enzymology , Animals , Signal Transduction
12.
Int J Mol Sci ; 25(13)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-39000336

ABSTRACT

Neurodegenerative diseases affect an increasing part of the population of modern societies, burdening healthcare systems and causing immense suffering at the personal level. The pathogenesis of several of these disorders involves dysregulation of gene expression, which depends on several molecular processes ranging from transcription to protein stability. microRNAs (miRNAs) are short non-coding RNA molecules that modulate gene expression by suppressing the translation of partially complementary mRNAs. miR-137 is a conserved, neuronally enriched miRNA that is implicated in neurodegeneration. Here, we review the current body of knowledge about the role that miR-137 plays in five prominent neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and multiple sclerosis. The presented data indicate that, rather than having a general neuroprotective role, miR-137 modulates the pathology of distinct disorders differently.


Subject(s)
MicroRNAs , Neurodegenerative Diseases , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Animals , Gene Expression Regulation
13.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39000479

ABSTRACT

It has been widely established that the characterization of extracellular vesicles (EVs), particularly small EVs (sEVs), shed by different cell types into biofluids, helps to identify biomarkers and therapeutic targets in neurological and neurodegenerative diseases. Recent studies are also exploring the efficacy of mesenchymal stem cell-derived extracellular vesicles naturally enriched with therapeutic microRNAs and proteins for treating various diseases. In addition, EVs released by various neural cells play a crucial function in the modulation of signal transmission in the brain in physiological conditions. However, in pathological conditions, such EVs can facilitate the spread of pathological proteins from one brain region to the other. On the other hand, the analysis of EVs in biofluids can identify sensitive biomarkers for diagnosis, prognosis, and disease progression. This review discusses the potential therapeutic use of stem cell-derived EVs in several central nervous system diseases. It lists their differences and similarities and confers various studies exploring EVs as biomarkers. Further advances in EV research in the coming years will likely lead to the routine use of EVs in therapeutic settings.


Subject(s)
Biomarkers , Central Nervous System Diseases , Extracellular Vesicles , Humans , Extracellular Vesicles/metabolism , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/therapy , Central Nervous System Diseases/diagnosis , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Mesenchymal Stem Cells/metabolism , Neurodegenerative Diseases/therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/diagnosis
14.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-39000543

ABSTRACT

Human individual differences in brain cytochrome P450 (CYP) metabolism, including induction, inhibition, and genetic variation, may influence brain sensitivity to neurotoxins and thus participate in the onset of neurodegenerative diseases. The aim of this study was to explore the modulation of CYPs in neuronal cells. The experimental approach was focused on differentiating human neuroblastoma SH-SY5Y cells into a phenotype resembling mature dopamine neurons and investigating the effects of specific CYP isoform induction. The results demonstrated that the differentiation protocols using retinoic acid followed by phorbol esters or brain-derived neurotrophic factor successfully generated SH-SY5Y cells with morphological neuronal characteristics and increased neuronal markers (NeuN, synaptophysin, ß-tubulin III, and MAO-B). qRT-PCR and Western blot analysis showed that expression of the CYP 1A1, 3A4, 2D6, and 2E1 isoforms was detectable in undifferentiated cells, with subsequent increases in CYP 2E1, 2D6, and 1A1 following differentiation. Further increases in the 1A1, 2D6, and 2E1 isoforms following ß-naphthoflavone treatment and 1A1 and 2D6 isoforms following ethanol treatment were evident. These results demonstrate that CYP isoforms can be modulated in SH-SY5Y cells and suggest their potential as an experimental model to investigate the role of CYPs in neuronal processes involved in the development of neurodegenerative diseases.


Subject(s)
Cell Differentiation , Cytochrome P-450 Enzyme System , Neurodegenerative Diseases , Humans , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/genetics , Cell Line, Tumor , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Tretinoin/pharmacology , Tretinoin/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neuroblastoma/genetics , Isoenzymes/metabolism , Isoenzymes/genetics , Dopaminergic Neurons/metabolism , Neurons/metabolism
15.
Neuropathol Appl Neurobiol ; 50(4): e12997, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39010256

ABSTRACT

AIMS: Recent advances in artificial intelligence, particularly with large language models like GPT-4Vision (GPT-4V)-a derivative feature of ChatGPT-have expanded the potential for medical image interpretation. This study evaluates the accuracy of GPT-4V in image classification tasks of histopathological images and compares its performance with a traditional convolutional neural network (CNN). METHODS: We utilised 1520 images, including haematoxylin and eosin staining and tau immunohistochemistry, from patients with various neurodegenerative diseases, such as Alzheimer's disease (AD), progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD). We assessed GPT-4V's performance using multi-step prompts to determine how textual context influences image interpretation. We also employed few-shot learning to enhance improvements in GPT-4V's diagnostic performance in classifying three specific tau lesions-astrocytic plaques, neuritic plaques and tufted astrocytes-and compared the outcomes with the CNN model YOLOv8. RESULTS: GPT-4V accurately recognised staining techniques and tissue origin but struggled with specific lesion identification. The interpretation of images was notably influenced by the provided textual context, which sometimes led to diagnostic inaccuracies. For instance, when presented with images of the motor cortex, the diagnosis shifted inappropriately from AD to CBD or PSP. However, few-shot learning markedly improved GPT-4V's diagnostic capabilities, enhancing accuracy from 40% in zero-shot learning to 90% with 20-shot learning, matching the performance of YOLOv8, which required 100-shot learning to achieve the same accuracy. CONCLUSIONS: Although GPT-4V faces challenges in independently interpreting histopathological images, few-shot learning significantly improves its performance. This approach is especially promising for neuropathology, where acquiring extensive labelled datasets is often challenging.


Subject(s)
Neural Networks, Computer , Neurodegenerative Diseases , Humans , Neurodegenerative Diseases/pathology , Image Interpretation, Computer-Assisted/methods , Alzheimer Disease/pathology
16.
Mech Ageing Dev ; 220: 111960, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38971236

ABSTRACT

Neurodegenerative diseases have multifactorial pathogenesis, mainly involving neuroinflammatory processes. Finding drugs able to treat these diseases, expecially because for most of these diseases there are no effective drugs, and the current drugs cause undesired side effects, represent a crucial point. Most in vivo and in vitro studies have been concentrated on various aspects related to neurons (e.g. neuroprotection), however, there has not been focus on the prevention of early stages involving glial cell activation and neuroinflammation. Recently, it has been demonstrated that nutritional phytochemicals including polyphenols, the main active constituents of the Mediterranean diet, maintain redox balance and neuroprotection through the activation of hormetic vitagene pathway. Recent lipidomics data from our laboratory indicate mushrooms as strong nutritional neuronutrients with strongly activity against neuroinflammation in Meniere' diseaseas, a model of cochleovestibular neural degeneration, as well as in animal model of traumatic brain injury, or rotenone induced parkinson's disease. Moreover, Hidrox®, an aqueous extract of olive containing hydroxytyrosol, and Boswellia, acting as Nrf2 activators, promote resilience by enhancing the redox potential, and thus, regulate through hormetic mechanisms, cellular stress response mechanisms., Thus, modulation of cellular stress pathways, in particular vitagenes system, may be an innovative approach for therapeutic intervention in neurodegenerative disorders.


Subject(s)
Hormesis , Humans , Animals , Healthy Aging/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/drug therapy
17.
J Am Heart Assoc ; 13(14): e034225, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38979810

ABSTRACT

BACKGROUND: The purpose of this study was to investigate the relationship between blood-brain barrier (BBB) permeability and cognitive functioning in healthy older adults and individuals with neurodegenerative diseases. METHODS AND RESULTS: A total of 124 participants with Alzheimer disease, cerebrovascular disease, or a mix Alzheimer's and cerebrovascular diseases and 55 controlparticipants underwent magnetic resonance imaging and neuropsychological testing. BBB permeability was measured with dynamic contrast-enhanced magnetic resonance imaging and white matter injury was measured using a quantitative diffusion-tensor imaging marker of white matter injury. Structural equation modeling was used to examine the relationships between BBB permeability, vascular risk burden, white matter injury, and cognitive functioning. Vascular risk burden predicted BBB permeability (r=0.24, P<0.05) and white matter injury (r=0.38, P<0.001). BBB permeability predicted increased white matter injury (r=0.34, P<0.001) and increased white matter injury predicted lower cognitive functioning (r=-0.51, P<0.001). CONCLUSIONS: The study provides empirical support for a vascular contribution to white matter injury and cognitive impairment, directly or indirectly via BBB permeability. This highlights the importance of targeting modifiable vascular risk factors to help mitigate future cognitive decline.


Subject(s)
Blood-Brain Barrier , Cognition , Humans , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Male , Female , Aged , Cognition/physiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Capillary Permeability , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/etiology , Middle Aged , Aged, 80 and over , White Matter/diagnostic imaging , White Matter/metabolism , White Matter/pathology , Neuropsychological Tests , Magnetic Resonance Imaging , Case-Control Studies , Diffusion Tensor Imaging , Aging/metabolism , Aging/psychology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Healthy Aging
18.
Cells ; 13(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38994964

ABSTRACT

Aggregation of the microtubule-associated protein tau (MAPT) is the hallmark pathology in a spectrum of neurodegenerative disorders collectively called tauopathies. Physiologically, tau is an inherent neuronal protein that plays an important role in the assembly of microtubules and axonal transport. However, disease-associated mutations of this protein reduce its binding to the microtubule components and promote self-aggregation, leading to formation of tangles in neurons. Tau is also expressed in oligodendrocytes, where it has significant developmental roles in oligodendrocyte maturation and myelin synthesis. Oligodendrocyte-specific tau pathology, in the form of fibrils and coiled coils, is evident in major tauopathies including progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and Pick's disease (PiD). Multiple animal models of tauopathy expressing mutant forms of MAPT recapitulate oligodendroglial tau inclusions with potential to cause degeneration/malfunction of oligodendrocytes and affecting the neuronal myelin sheath. Till now, mechanistic studies heavily concentrated on elucidating neuronal tau pathology. Therefore, more investigations are warranted to comprehensively address tau-induced pathologies in oligodendrocytes. The present review provides the current knowledge available in the literature about the intricate relations between tau and oligodendrocytes in health and diseases.


Subject(s)
Oligodendroglia , Tauopathies , tau Proteins , Humans , Tauopathies/metabolism , Tauopathies/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Animals , tau Proteins/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology
19.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-39000326

ABSTRACT

Decades of research have identified genetic and environmental factors involved in age-related neurodegenerative diseases and, to a lesser extent, neuropsychiatric disorders. Genomic instability, i.e., the loss of genome integrity, is a common feature among both neurodegenerative (mayo-trophic lateral sclerosis, Parkinson's disease, Alzheimer's disease) and psychiatric (schizophrenia, autism, bipolar depression) disorders. Genomic instability is associated with the accumulation of persistent DNA damage and the activation of DNA damage response (DDR) pathways, as well as pathologic neuronal cell loss or senescence. Typically, DDR signaling ensures that genomic and proteomic homeostasis are maintained in both dividing cells, including neural progenitors, and post-mitotic neurons. However, dysregulation of these protective responses, in part due to aging or environmental insults, contributes to the progressive development of neurodegenerative and/or psychiatric disorders. In this Special Issue, we introduce and highlight the overlap between neurodegenerative diseases and neuropsychiatric disorders, as well as the emerging clinical, genomic, and molecular evidence for the contributions of DNA damage and aberrant DNA repair. Our goal is to illuminate the importance of this subject to uncover possible treatment and prevention strategies for relevant devastating brain diseases.


Subject(s)
DNA Damage , Genomic Instability , Mental Disorders , Neurodegenerative Diseases , Animals , Humans , DNA Repair , Mental Disorders/metabolism , Mental Disorders/etiology , Mental Disorders/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/genetics
20.
Open Biol ; 14(7): 230419, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39013416

ABSTRACT

The mechanisms responsible for neuronal death causing cognitive loss in Alzheimer's disease (AD) and many other dementias are not known. Serum amyloid P component (SAP) is a constitutive plasma protein, which is cytotoxic for cerebral neurones and also promotes formation and persistence of cerebral Aß amyloid and neurofibrillary tangles. Circulating SAP, which is produced exclusively by the liver, is normally almost completely excluded from the brain. Conditions increasing brain exposure to SAP increase dementia risk, consistent with a causative role in neurodegeneration. Furthermore, neocortex content of SAP is strongly and independently associated with dementia at death. Here, seeking genomic evidence for a causal link of SAP with neurodegeneration, we meta-analysed three genome-wide association studies of 44 288 participants, then conducted cis-Mendelian randomization assessment of associations with neurodegenerative diseases. Higher genetically instrumented plasma SAP concentrations were associated with AD (odds ratio 1.07, 95% confidence interval (CI) 1.02; 1.11, p = 1.8 × 10-3), Lewy body dementia (odds ratio 1.37, 95%CI 1.19; 1.59, p = 1.5 × 10-5) and plasma tau concentration (0.06 log2(ng l-1) 95%CI 0.03; 0.08, p = 4.55 × 10-6). These genetic findings are consistent with neuropathogenicity of SAP. Depletion of SAP from the blood and the brain, by the safe, well tolerated, experimental drug miridesap may thus be neuroprotective.


Subject(s)
Genome-Wide Association Study , Neurodegenerative Diseases , Serum Amyloid P-Component , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Serum Amyloid P-Component/metabolism , Serum Amyloid P-Component/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/etiology , Polymorphism, Single Nucleotide , Genetic Predisposition to Disease , Mendelian Randomization Analysis , Biomarkers , tau Proteins/metabolism , tau Proteins/genetics , Lewy Body Disease/genetics , Lewy Body Disease/metabolism , Male , Female
SELECTION OF CITATIONS
SEARCH DETAIL
...