Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29.235
Filtrar
1.
Results Probl Cell Differ ; 73: 203-227, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39242381

RESUMEN

Tunneling nanotubes (TNTs) have emerged as intriguing structures facilitating intercellular communications across diverse cell types, which are integral to several biological processes, as well as participating in various disease progression. This review provides an in-depth analysis of TNTs, elucidating their structural characteristics and functional roles, with a particular focus on their significance within the brain environment and their implications in neurological and neurodegenerative disorders. We explore the interplay between TNTs and neurological diseases, offering potential mechanistic insights into disease progression, while also highlighting their potential as viable therapeutic targets. Additionally, we address the significant challenges associated with studying TNTs, from technical limitations to their investigation in complex biological systems. By addressing some of these challenges, this review aims to pave the way for further exploration into TNTs, establishing them as a central focus in advancing our understanding of neurodegenerative disorders.


Asunto(s)
Encéfalo , Comunicación Celular , Enfermedades Neurodegenerativas , Humanos , Encéfalo/fisiología , Animales , Comunicación Celular/fisiología , Nanotubos/química , Estructuras de la Membrana Celular
2.
Brain Nerve ; 76(9): 1059-1064, 2024 Sep.
Artículo en Japonés | MEDLINE | ID: mdl-39251226

RESUMEN

Neurodegenerative diseases represent the most common cause of dementia. Protein aggregation is upstream in the pathological mechanisms and is a therapeutic target in the development of disease-modifying drugs in this patient population. Notably, α-synuclein or DNA-binding protein of 43kDa (TDP-43) is commonly involved in the pathomechanisms that contribute to non-Alzheimer neurodegenerative diseases. Several immunotherapy clinical trials on α-synuclein have progressed to phase 2, and small-molecule therapeutics are ongoing. With regard to TDP-43, immunotherapies that target protein aggregates are currently being developed, and research is underway to investigate several drugs that target the associated causative gene. Further research is warranted for deeper insight into both disease-modifying drugs; biomarker tests need to be developed to determine their efficacy. However, both proteins aggregate and accumulate in the brain in many neurodegenerative diseases and dementia; therefore, they are therapeutically significant, and future progress is expected in research and development.


Asunto(s)
Demencia , Humanos , Demencia/tratamiento farmacológico , alfa-Sinucleína/metabolismo , Proteínas de Unión al ADN/metabolismo , Inmunoterapia , Enfermedades Neurodegenerativas/tratamiento farmacológico
3.
Traffic ; 25(9): e12951, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39238078

RESUMEN

Mitochondria, the dynamic organelles responsible for energy production and cellular metabolism, have the metabolic function of extracting energy from nutrients and synthesizing crucial metabolites. Nevertheless, recent research unveils that intercellular mitochondrial transfer by tunneling nanotubes, tumor microtubes, gap junction intercellular communication, extracellular vesicles, endocytosis and cell fusion may regulate mitochondrial function within recipient cells, potentially contributing to disease treatment, such as nonalcoholic steatohepatitis, glioblastoma, ischemic stroke, bladder cancer and neurodegenerative diseases. This review introduces the principal approaches to intercellular mitochondrial transfer and examines its role in various diseases. Furthermore, we provide a comprehensive overview of the inhibitors and activators of intercellular mitochondrial transfer, offering a unique perspective to illustrate the relationship between intercellular mitochondrial transfer and diseases.


Asunto(s)
Mitocondrias , Humanos , Mitocondrias/metabolismo , Animales , Comunicación Celular , Vesículas Extracelulares/metabolismo , Transporte Biológico , Endocitosis/fisiología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/terapia
4.
Front Immunol ; 15: 1426656, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39238639

RESUMEN

Neurodegenerative diseases (NDs) in mammals, such as Alzheimer's disease (AD), Parkinson's disease (PD), and transmissible spongiform encephalopathies (TSEs), are characterized by the accumulation of misfolded proteins in the central nervous system (CNS). Despite the presence of these pathogenic proteins, the immune response in affected individuals remains notably muted. Traditional immunological strategies, particularly those reliant on monoclonal antibodies (mAbs), face challenges related to tissue penetration, blood-brain barrier (BBB) crossing, and maintaining protein stability. This has led to a burgeoning interest in alternative immunotherapeutic avenues. Notably, single-domain antibodies (or nanobodies) and aptamers have emerged as promising candidates, as their reduced size facilitates high affinity antigen binding and they exhibit superior biophysical stability compared to mAbs. Aptamers, synthetic molecules generated from DNA or RNA ligands, present both rapid production times and cost-effective solutions. Both nanobodies and aptamers exhibit inherent qualities suitable for ND research and therapeutic development. Cross-seeding events must be considered in both traditional and small-molecule-based immunodiagnostic and therapeutic approaches, as well as subsequent neurotoxic impacts and complications beyond protein aggregates. This review delineates the challenges traditional immunological methods pose in ND research and underscores the potential of nanobodies and aptamers in advancing next-generation ND diagnostics and therapeutics.


Asunto(s)
Aptámeros de Nucleótidos , Enfermedades Neurodegenerativas , Anticuerpos de Dominio Único , Humanos , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/uso terapéutico , Aptámeros de Nucleótidos/uso terapéutico , Aptámeros de Nucleótidos/inmunología , Animales , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/terapia
5.
Transl Neurodegener ; 13(1): 46, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39242576

RESUMEN

Neurodegenerative disorders are typically "split" based on their hallmark clinical, anatomical, and pathological features, but they can also be "lumped" by a shared feature of impaired mitochondrial biology. This leads us to present a scientific framework that conceptualizes Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD) as "metabolic icebergs" comprised of a tip, a bulk, and a base. The visible tip conveys the hallmark neurological symptoms, neurodegenerative regions, and neuronal protein aggregates for each disorder. The hidden bulk depicts impaired mitochondrial biology throughout the body, which is multifaceted and may be subdivided into impaired cellular metabolism, cell-specific mitotypes, and mitochondrial behaviours, functions, activities, and features. The underlying base encompasses environmental factors, especially modern industrial toxins, dietary lifestyles, and cognitive, physical, and psychosocial behaviours, but also accommodates genetic factors specific to familial forms of AD, PD, and ALS, as well as HD. Over years or decades, chronic exposure to a particular suite of environmental and genetic factors at the base elicits a trajectory of impaired mitochondrial biology that maximally impacts particular subsets of mitotypes in the bulk, which eventually surfaces as the hallmark features of a particular neurodegenerative disorder at the tip. We propose that impaired mitochondrial biology can be repaired and recalibrated by activating "mitohormesis", which is optimally achieved using strategies that facilitate a balanced oscillation between mitochondrial stressor and recovery phases. Sustainably harnessing mitohormesis may constitute a potent preventative and therapeutic measure for people at risk of, or suffering with, neurodegenerative disorders.


Asunto(s)
Mitocondrias , Enfermedades Neurodegenerativas , Humanos , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/genética , Mitocondrias/metabolismo , Hormesis/fisiología , Animales
6.
Cell Mol Biol Lett ; 29(1): 116, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39237893

RESUMEN

Lysosomes are acidic organelles involved in crucial intracellular functions, including the degradation of organelles and protein, membrane repair, phagocytosis, endocytosis, and nutrient sensing. Given these key roles of lysosomes, maintaining their homeostasis is essential for cell viability. Thus, to preserve lysosome integrity and functionality, cells have developed a complex intracellular system, called lysosome quality control (LQC). Several stressors may affect the integrity of lysosomes, causing Lysosomal membrane permeabilization (LMP), in which membrane rupture results in the leakage of luminal hydrolase enzymes into the cytosol. After sensing the damage, LQC either activates lysosome repair, or induces the degradation of the ruptured lysosomes through autophagy. In addition, LQC stimulates the de novo biogenesis of functional lysosomes and lysosome exocytosis. Alterations in LQC give rise to deleterious consequences for cellular homeostasis. Specifically, the persistence of impaired lysosomes or the malfunctioning of lysosomal processes leads to cellular toxicity and death, thereby contributing to the pathogenesis of different disorders, including neurodegenerative diseases (NDs). Recently, several pieces of evidence have underlined the importance of the role of lysosomes in NDs. In this review, we describe the elements of the LQC system, how they cooperate to maintain lysosome homeostasis, and their implication in the pathogenesis of different NDs.


Asunto(s)
Lisosomas , Enfermedades Neurodegenerativas , Lisosomas/metabolismo , Humanos , Enfermedades Neurodegenerativas/metabolismo , Animales , Homeostasis , Autofagia
7.
Acta Biochim Biophys Sin (Shanghai) ; 56(8): 1208-1220, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39225075

RESUMEN

Aging, a complex biological process, involves the progressive decline of physiological functions across various systems, leading to increased susceptibility to neurodegenerative diseases. In society, demographic aging imposes significant economic and social burdens due to these conditions. This review specifically examines the association of protein glycosylation with aging and neurodegenerative diseases. Glycosylation, a critical post-translational modification, influences numerous aspects of protein function that are pivotal in aging and the pathophysiology of diseases such as Alzheimer's disease, Parkinson's disease, and other neurodegenerative conditions. We highlight the alterations in glycosylation patterns observed during aging, their implications in the onset and progression of neurodegenerative diseases, and the potential of glycosylation profiles as biomarkers for early detection, prognosis, and monitoring of these age-associated conditions, and delve into the mechanisms of glycosylation. Furthermore, this review explores their role in regulating protein function and mediating critical biological interactions in these diseases. By examining the changes in glycosylation profiles associated with each part, this review underscores the potential of glycosylation research as a tool to enhance our understanding of aging and its related diseases.


Asunto(s)
Envejecimiento , Enfermedades Neurodegenerativas , Procesamiento Proteico-Postraduccional , Humanos , Glicosilación , Envejecimiento/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Biomarcadores/metabolismo , Enfermedad de Parkinson/metabolismo
8.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(9): 1131-1138, 2024 Sep 10.
Artículo en Chino | MEDLINE | ID: mdl-39217495

RESUMEN

Polyglutamine (PolyQ) disease is a group of hereditary neurodegenerative diseases. It has become clear that brain damage may occur decades before the onset of symptoms. As a sensitive marker for neuro-axonal damages, neurofilament light chain (NfL) has appeared as a promising biomarker for neurological diseases. it may be used as a preclinical and clinical marker for the neurodegeneration in polyQ diseases, and is closely correlated with disease severity and progression, in particular different disease stages. This article has provided a review for the value of NfL as a biomarker in polyQ disease and its future research directions.


Asunto(s)
Enfermedades Neurodegenerativas , Proteínas de Neurofilamentos , Péptidos , Humanos , Péptidos/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Proteínas de Neurofilamentos/genética , Biomarcadores , Animales
9.
Artículo en Inglés | MEDLINE | ID: mdl-39284324

RESUMEN

With the accelerated aging of the global population, the incidence of neurodegenerative diseases (NDDs) is increasing year by year. Because of the presence of the blood-brain barrier (BBB), the low concentration of the biomarkers in peripheral blood and the low penetration rate of the drugs through BBB into brain hinders the development of diagnosis and treatment of NDDs. As an effective mediator to penetrate through BBB in both directions, extracellular vesicles (EVs) have attracted much attention in the early diagnosis and treatment of NDDs because of their superior performance as drug carriers and detection biomarkers. Brain-derived EVs in body fluids contain disease-related biomolecules can be used as early diagnostic biomarkers for NDDs. In addition, as one of the subpopulations of EVs, exosomes, especially stem cell-derived exosomes, have great potential in the treatment of NDDs. The ability to cross the BBB, together with the feasibility of versatile functionalization of EV for NDDs pathogen targeting facilitate EVs a potential tool for targeted drug delivery systems for NDDs. In this review, the important role of EVs in the diagnosis and treatment of NDDs and the current research progress will be discussed. This article is categorized under: Diagnostic Tools > Diagnostic Nanodevices Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease.


Asunto(s)
Barrera Hematoencefálica , Vesículas Extracelulares , Enfermedades Neurodegenerativas , Nanomedicina Teranóstica , Humanos , Enfermedades Neurodegenerativas/diagnóstico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Vesículas Extracelulares/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Biomarcadores/metabolismo , Sistemas de Liberación de Medicamentos , Ratones
10.
AAPS PharmSciTech ; 25(7): 215, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39266806

RESUMEN

Neurodegenerative disorder refers to malfunctioning of neurons their degradation leading to death of neurons. Among various neurodegenerative disorders APHD (Alzheimer's, Parkinson's, and Huntington's Disease) are particularly concerning due to their progressive and debilitating nature. The therapeutic agent used for treatment and management of APHD often show unsatisfactory clinical outcome owing to poor solubility and limited permeability across blood brain barrier (BBB). The nose-to brain delivery can overcome this BBB challenge as it can transport drug directly to brain though olfactory pathways bypassing BBB. Additionally, the nanotechnology has emerged as a cutting-edge methodology to address this issue and specifically mucoadhesive micro/nanoemulsion can improve the overall performance of the drug when administered intranasally. Beyond the therapy neurotechnology has emerged as are revolutionary AI-driven BCI (Brain computer interface) aimed to restore independence in patients with function loss due to neuron degeneration/death. A promising BCI Neuralink has been recently explored for clinical trials and results revealed that a quadriplegia bearing person with implanted Neuralink chip was able to perform few normal functions of daily routine such as playing online games, text messaging, reading, and learning foreign languages online through accessing the particular websites. This review will discuss the fundamental concepts of neurodegeneration, application of micro/nanoemulsion through intranasal route and integration of neurotechnology for the management and treatment of APHD.


Asunto(s)
Administración Intranasal , Sistemas de Liberación de Medicamentos , Emulsiones , Nanotecnología , Enfermedades Neurodegenerativas , Administración Intranasal/métodos , Humanos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Nanotecnología/métodos , Animales , Barrera Hematoencefálica/metabolismo , Nanopartículas/química , Nanopartículas/administración & dosificación
11.
Cells ; 13(17)2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39273013

RESUMEN

Vacuolar-type ATPase (v-ATPase) is a multimeric protein complex that regulates H+ transport across membranes and intra-cellular organelle acidification. Catabolic processes, such as endocytic degradation and autophagy, strictly rely on v-ATPase-dependent luminal acidification in lysosomes. The v-ATPase complex is expressed at high levels in the brain and its impairment triggers neuronal dysfunction and neurodegeneration. Due to their post-mitotic nature and highly specialized function and morphology, neurons display a unique vulnerability to lysosomal dyshomeostasis. Alterations in genes encoding subunits composing v-ATPase or v-ATPase-related proteins impair brain development and synaptic function in animal models and underlie genetic diseases in humans, such as encephalopathies, epilepsy, as well as neurodevelopmental, and degenerative disorders. This review presents the genetic and functional evidence linking v-ATPase subunits and accessory proteins to various brain disorders, from early-onset developmental epileptic encephalopathy to neurodegenerative diseases. We highlight the latest emerging therapeutic strategies aimed at mitigating lysosomal defects associated with v-ATPase dysfunction.


Asunto(s)
Encéfalo , ATPasas de Translocación de Protón Vacuolares , Humanos , ATPasas de Translocación de Protón Vacuolares/metabolismo , ATPasas de Translocación de Protón Vacuolares/genética , Encéfalo/patología , Encéfalo/metabolismo , Animales , Lisosomas/metabolismo , Lisosomas/enzimología , Encefalopatías/genética , Encefalopatías/metabolismo , Encefalopatías/enzimología , Encefalopatías/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo
12.
Int J Mol Sci ; 25(17)2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39273288

RESUMEN

Cellular metabolism is crucial for various physiological processes, with folate-dependent one-carbon (1C) metabolism playing a pivotal role. Folate, a B vitamin, is a key cofactor in this pathway, supporting DNA synthesis, methylation processes, and antioxidant defenses. In dividing cells, folate facilitates nucleotide biosynthesis, ensuring genomic stability and preventing carcinogenesis. Additionally, in neurodevelopment, folate is essential for neural tube closure and central nervous system formation. Thus, dysregulation of folate metabolism can contribute to pathologies such as cancer, severe birth defects, and neurodegenerative diseases. Epidemiological evidence highlights folate's impact on disease risk and its potential as a therapeutic target. In cancer, antifolate drugs that inhibit key enzymes of folate-dependent 1C metabolism and strategies targeting folate receptors are current therapeutic options. However, folate's impact on cancer risk is complex, varying among cancer types and dietary contexts. In neurodegenerative conditions, including Alzheimer's and Parkinson's diseases, folate deficiency exacerbates cognitive decline through elevated homocysteine levels, contributing to neuronal damage. Clinical trials of folic acid supplementation show mixed outcomes, underscoring the complexities of its neuroprotective effects. This review integrates current knowledge on folate metabolism in cancer and neurodegeneration, exploring molecular mechanisms, clinical implications, and therapeutic strategies, which can provide crucial information for advancing treatments.


Asunto(s)
Ácido Fólico , Neoplasias , Enfermedades Neurodegenerativas , Humanos , Ácido Fólico/metabolismo , Ácido Fólico/uso terapéutico , Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Animales , Carbono/metabolismo , Antagonistas del Ácido Fólico/uso terapéutico , Antagonistas del Ácido Fólico/farmacología
13.
Int J Mol Sci ; 25(17)2024 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-39273435

RESUMEN

Neurodegenerative disorders are the main cause of cognitive and physical disabilities, affect millions of people worldwide, and their incidence is on the rise. Emerging evidence pinpoints a disturbance of the communication of the gut-brain axis, and in particular to gut microbial dysbiosis, as one of the contributors to the pathogenesis of these diseases. In fact, dysbiosis has been associated with neuro-inflammatory processes, hyperactivation of the neuronal immune system, impaired cognitive functions, aging, depression, sleeping disorders, and anxiety. With the rapid advance in metagenomics, metabolomics, and big data analysis, together with a multidisciplinary approach, a new horizon has just emerged in the fields of translational neurodegenerative disease. In fact, recent studies focusing on taxonomic profiling and leaky gut in the pathogenesis of neurodegenerative disorders are not only shedding light on an overlooked field but are also creating opportunities for biomarker discovery and development of new therapeutic and adjuvant strategies to treat these disorders. Lactiplantibacillus plantarum (LBP) strains are emerging as promising psychobiotics for the treatment of these diseases. In fact, LBP strains are able to promote eubiosis, increase the enrichment of bacteria producing beneficial metabolites such as short-chain fatty acids, boost the production of neurotransmitters, and support the homeostasis of the gut-brain axis. In this review, we summarize the current knowledge on the role of the gut microbiota in the pathogenesis of neurodegenerative disorders with a particular focus on the benefits of LBP strains in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, autism, anxiety, and depression.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Neurodegenerativas , Probióticos , Humanos , Enfermedades Neurodegenerativas/microbiología , Enfermedades Neurodegenerativas/metabolismo , Probióticos/uso terapéutico , Disbiosis/microbiología , Eje Cerebro-Intestino , Animales
14.
Int J Mol Sci ; 25(17)2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39273460

RESUMEN

Degenerative diseases oftentimes occur within the continuous process of aging, and the corresponding clinical manifestations may be neurodegeneration, neoplastic diseases, or various human complex diseases. DNA methylation provides the opportunity to explore aging and degenerative diseases as epigenetic traits. It has already been applied to age prediction and disease diagnosis. It has been shown that various degenerative diseases share co-physiology mechanisms with each other, clues of which may be gained from studying the aging process. Here, we endeavor to predict the risk of degenerative diseases in an aging-relevant comorbid mechanism perspective. Firstly, an epigenetic clock method was implemented based on a multi-scale convolutional neural network, and a Shapley feature attribution analysis was applied to discover the aging-related CpG sites. Then, these sites were further screened to a smaller subset composed of 196 sites by using biomics analysis according to their biological functions and mechanisms. Finally, we constructed a multilayer perceptron (MLP)-based degenerative disease risk prediction model, Mlp-DDR, which was well trained and tested to accurately classify nine degenerative diseases. Recent studies also suggest that DNA methylation plays a significant role in conditions like osteoporosis and osteoarthritis, broadening the potential applications of our model. This approach significantly advances the ability to understand degenerative diseases and represents a substantial shift from traditional diagnostic methods. Despite the promising results, limitations regarding model complexity and dataset diversity suggest directions for future research, including the development of tissue-specific epigenetic clocks and the inclusion of a wider range of diseases.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Enfermedades Neurodegenerativas , Humanos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/diagnóstico , Islas de CpG , Envejecimiento/genética , Redes Neurales de la Computación
15.
Int J Mol Sci ; 25(17)2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39273694

RESUMEN

Experimental models play a pivotal role in biomedical research, facilitating the understanding of disease mechanisms and the development of novel therapeutics. This is particularly true for neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and motor neuron disease, which present complex challenges for research and therapy development. In this work, we review the recent literature about experimental models and motor neuron disease. We identified three main categories of models that are highly studied by scientists. In fact, experimental models for investigating these diseases encompass a variety of approaches, including modeling the patient's cell culture, patient-derived induced pluripotent stem cells, and organoids. Each model offers unique advantages and limitations, providing researchers with a range of tools to address complex biological questions. Here, we discuss the characteristics, applications, and recent advancements in terms of each model system, highlighting their contributions to advancing biomedical knowledge and translational research.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedades Neurodegenerativas , Organoides , Humanos , Enfermedades Neurodegenerativas/terapia , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/metabolismo , Animales , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Organoides/patología , Modelos Biológicos
16.
Molecules ; 29(17)2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39274839

RESUMEN

Tropomyosin kinase receptor B (TrkB) has been explored as a therapeutic target for neurological and psychiatric disorders. However, the development of TrkB agonists was hindered by our poor understanding of the TrkB agonist binding location and affinity (both affect the regulation of disorder types). This motivated us to develop a combined computational and experimental approach to study TrkB binders. First, we developed a docking method to simulate the binding affinity of TrkB and binders identified by our magnetic drug screening platform from Gotu kola extracts. The Fred Docking scores from the docking computation showed strong agreement with the experimental results. Subsequently, using this screening platform, we identified a list of compounds from the NIH clinical collection library and applied the same docking studies. From the Fred Docking scores, we selected two compounds for TrkB activation tests. Interestingly, the ability of the compounds to increase dendritic arborization in hippocampal neurons matched well with the computational results. Finally, we performed a detailed binding analysis of the top candidates and compared them with the best-characterized TrkB agonist, 7,8-dyhydroxyflavon. The screening platform directly identifies TrkB binders, and the computational approach allows for the quick selection of top candidates with potential biological activities based on the docking scores.


Asunto(s)
Simulación del Acoplamiento Molecular , Enfermedades Neurodegenerativas , Unión Proteica , Receptor trkB , Receptor trkB/metabolismo , Receptor trkB/agonistas , Humanos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Animales , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/agonistas
17.
Signal Transduct Target Ther ; 9(1): 238, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39256355

RESUMEN

Sex characteristics exhibit significant disparities in various human diseases, including prevalent cardiovascular diseases, cancers, metabolic disorders, autoimmune diseases, and neurodegenerative diseases. Risk profiles and pathological manifestations of these diseases exhibit notable variations between sexes. The underlying reasons for these sex disparities encompass multifactorial elements, such as physiology, genetics, and environment. Recent studies have shown that human body systems demonstrate sex-specific gene expression during critical developmental stages and gene editing processes. These genes, differentially expressed based on different sex, may be regulated by androgen or estrogen-responsive elements, thereby influencing the incidence and presentation of cardiovascular, oncological, metabolic, immune, and neurological diseases across sexes. However, despite the existence of sex differences in patients with human diseases, treatment guidelines predominantly rely on male data due to the underrepresentation of women in clinical trials. At present, there exists a substantial knowledge gap concerning sex-specific mechanisms and clinical treatments for diverse diseases. Therefore, this review aims to elucidate the advances of sex differences on human diseases by examining epidemiological factors, pathogenesis, and innovative progress of clinical treatments in accordance with the distinctive risk characteristics of each disease and provide a new theoretical and practical basis for further optimizing individualized treatment and improving patient prognosis.


Asunto(s)
Caracteres Sexuales , Humanos , Femenino , Masculino , Neoplasias/genética , Neoplasias/patología , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/patología , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/inmunología , Factores Sexuales , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología
18.
Commun Biol ; 7(1): 1103, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39251807

RESUMEN

Neurofilament light chain (NfL) levels in circulation have been established as a sensitive biomarker of neuro-axonal damage across a range of neurodegenerative disorders. Elucidation of the genetic architecture of blood NfL levels could provide new insights into molecular mechanisms underlying neurodegenerative disorders. In this meta-analysis of genome-wide association studies (GWAS) of blood NfL levels from eleven cohorts of European ancestry, we identify two genome-wide significant loci at 16p12 (UMOD) and 17q24 (SLC39A11). We observe association of three loci at 1q43 (FMN2), 12q14, and 12q21 with blood NfL levels in the meta-analysis of African-American ancestry. In the trans-ethnic meta-analysis, we identify three additional genome-wide significant loci at 1p32 (FGGY), 6q14 (TBX18), and 4q21. In the post-GWAS analyses, we observe the association of higher NfL polygenic risk score with increased plasma levels of total-tau, Aß-40, Aß-42, and higher incidence of Alzheimer's disease in the Rotterdam Study. Furthermore, Mendelian randomization analysis results suggest that a lower kidney function could cause higher blood NfL levels. This study uncovers multiple genetic loci of blood NfL levels, highlighting the genes related to molecular mechanism of neurodegeneration.


Asunto(s)
Estudio de Asociación del Genoma Completo , Enfermedades Neurodegenerativas , Proteínas de Neurofilamentos , Humanos , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/sangre , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/sangre , Predisposición Genética a la Enfermedad , Sitios Genéticos , Biomarcadores/sangre , Polimorfismo de Nucleótido Simple , Masculino , Femenino , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/sangre
19.
Commun Biol ; 7(1): 1107, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39251817

RESUMEN

The central nervous system (CNS) comprises a diverse range of brain cell types with distinct functions and gene expression profiles. Although single-cell RNA sequencing (scRNA-seq) provides new insights into the brain cell atlases, integrating large-scale CNS scRNA-seq data still encounters challenges due to the complexity and heterogeneity among CNS cell types/subtypes. In this study, we introduce a self-supervised contrastive learning method, called scCM, for integrating large-scale CNS scRNA-seq data. scCM brings functionally related cells close together while simultaneously pushing apart dissimilar cells by comparing the variations of gene expression, effectively revealing the heterogeneous relationships within the CNS cell types/subtypes. The effectiveness of scCM is evaluated on 20 CNS datasets covering 4 species and 10 CNS diseases. Leveraging these strengths, we successfully integrate the collected human CNS datasets into a large-scale reference to annotate cell types and subtypes in neural tissues. Results demonstrate that scCM provides an accurate annotation, along with rich spatial information of cell state. In summary, scCM is a robust and promising method for integrating large-scale CNS scRNA-seq data, enabling researchers to gain insights into the cellular and molecular mechanisms underlying CNS functions and diseases.


Asunto(s)
Sistema Nervioso Central , Análisis de Expresión Génica de una Sola Célula , Aprendizaje Automático Supervisado , Sistema Nervioso Central/citología , Humanos , Conjuntos de Datos como Asunto , Análisis por Conglomerados , Enfermedades Neurodegenerativas/patología , Atlas como Asunto , Animales , Aprendizaje Profundo
20.
BMJ Open Diabetes Res Care ; 12(4)2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39242121

RESUMEN

INTRODUCTION: Diabetes is linked to neurodegenerative diseases (NDs), but data in type 1 diabetes are scarce. Our aim was to assess the standardized incidence ratios (SIRs) of different NDs in type 1 diabetes, and to evaluate the impact of diabetic vascular complications and age at diabetes onset. RESEARCH DESIGN AND METHODS: In this observational cohort study, we included 4261 individuals with type 1 diabetes from the Finnish Diabetic Nephropathy study, and 11 653 matched population-based controls without diabetes. NDs were identified from registers until the end of 2017. Diabetic complications were assessed at the baseline study visit. SIRs were calculated from diabetes onset, except for impact of complications that was calculated from baseline study visit. RESULTS: The SIRs for NDs were increased in type 1 diabetes: any dementia 2.24 (95% CI 1.79 to 2.77), Alzheimer's disease 2.13 (95% CI 1.55 to 2.87), vascular dementia 3.40 (95% CI 2.08 to 5.6), other dementias 1.70 (95% CI 1.22 to 2.31), and Parkinson's disease 1.61 (95% CI 1.04 to 2.37). SIR showed a twofold increased incidence already in those without albuminuria (1.99 (1.44-2.68)), but further increased in presence of diabetic complications: kidney disease increased SIR for Alzheimer's disease, while cardiovascular disease increased SIR for both Alzheimer's disease and other dementias. Diabetes onset <15 years, compared with ≥15 years, increased SIR of Alzheimer's disease, 3.89 (2.21-6.35) vs 1.73 (1.16-2.48), p<0.05, but not the other dementias. CONCLUSIONS: ND incidence is increased 1.7-3.4-fold in type 1 diabetes. The presence of diabetic kidney disease and cardiovascular disease further increased the incidence of dementia.


Asunto(s)
Diabetes Mellitus Tipo 1 , Enfermedades Neurodegenerativas , Humanos , Diabetes Mellitus Tipo 1/epidemiología , Diabetes Mellitus Tipo 1/complicaciones , Finlandia/epidemiología , Masculino , Femenino , Incidencia , Enfermedades Neurodegenerativas/epidemiología , Enfermedades Neurodegenerativas/complicaciones , Persona de Mediana Edad , Adulto , Estudios de Seguimiento , Anciano , Estudios de Casos y Controles , Estudios de Cohortes , Factores de Riesgo , Nefropatías Diabéticas/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA