Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.710
Filtrar
1.
Neuron ; 112(19): 3223-3225, 2024 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-39389009

RESUMO

The diverse etiologies of the genetic neurodegenerative disorder known as primary familial brain calcification have dimmed hopes for curative therapies. However, two new papers in Neuron1,2 provide a reason for optimism by identifying mechanisms involved in brain phosphate transport and a promising target for restoring phosphate balance in the brain.


Assuntos
Calcinose , Humanos , Calcinose/genética , Calcinose/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Animais , Fosfatos/metabolismo , Encefalopatias/genética , Encefalopatias/terapia , Encefalopatias/metabolismo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/terapia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo
2.
Cell Biol Toxicol ; 40(1): 83, 2024 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-39367211

RESUMO

Exogenous gaseous formaldehyde (FA) is recognized as a significant indoor air pollutant due to its chemical reactivity and documented mutagenic and carcinogenic properties, particularly in its capacity to damage DNA and impact human health. Despite increasing attention on the adverse effects of exogenous FA on human health, the potential detrimental effects of endogenous FA in the brain have been largely neglected in current research. Endogenous FA have been observed to accumulate in the aging brain due to dysregulation in the expression and activity of enzymes involved in FA metabolism. Surprisingly, excessive FA have been implicated in the development of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and brain cancers. Notably, FA has the ability to not only initiate DNA double strand breaks but also induce the formation of crosslinks of DNA-DNA, DNA-RNA, and DNA-protein, which further exacerbate the progression of these brain diseases. However, recent research has identified that FA-resistant gene exonuclease-1 (EXO1) and FA scavengers can potentially mitigate FA toxicity, offering a promising strategy for mitigating or repairing FA-induced DNA damage. The present review offers novel insights into the impact of FA metabolism on brain ageing and the contribution of FA-damaged DNA to the progression of neurological disorders.


Assuntos
Envelhecimento , Encéfalo , Dano ao DNA , Formaldeído , Humanos , Formaldeído/toxicidade , Formaldeído/efeitos adversos , Envelhecimento/metabolismo , Envelhecimento/genética , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Dano ao DNA/efeitos dos fármacos , Animais , Encefalopatias/induzido quimicamente , Encefalopatias/metabolismo , Encefalopatias/patologia , Encefalopatias/genética
3.
Sci Rep ; 14(1): 24044, 2024 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-39402106

RESUMO

Diabetic encephalopathy (DE) is a severe complication of diabetes, but its pathogenesis remains unclear. This study aimed to investigate the roles and underlying mechanisms of high glucose (HG)- and advanced glycosylation end product (AGE)-induced oxidative stress (OS) in the cognitive decline in DE. The DE mouse model was established using a high-fat diet and streptozotocin, and its cognitive functions were evaluated using the Morris Water Maze, novel object recognition, and Y-maze test. The results revealed increased reactive oxygen species (ROS) generation, mitophagy inhibition, and decreased prohibitin 2 (PHB2) expression in the hippocampal neurons of DE mice and HG- or AGE-treated HT-22 cells. However, overexpression of PHB2 reduced ROS generation, reversed mitophagy inhibition, and improved mitochondrial function in the HG- or AGE-treated HT-22 cells and ameliorated cognitive decline, improved mitochondrial structural damage, and reversed mitophagy inhibition of hippocampal neurons in DE mice. Further analysis revealed that the Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor erythroid 2-related factor 2 (Nrf2) pathway was involved in the HG- or AGE-mediated downregulation of PHB2 in HT-22 cells. These results demonstrate that HG- or AGE-induced OS inhibits the mitophagy of hippocampal neurons via the Keap1-Nrf2-PHB2 pathway, thereby contributing to the cognitive decline in DE.


Assuntos
Glucose , Hipocampo , Proteína 1 Associada a ECH Semelhante a Kelch , Mitofagia , Fator 2 Relacionado a NF-E2 , Neurônios , Estresse Oxidativo , Proibitinas , Proteínas Repressoras , Animais , Hipocampo/metabolismo , Hipocampo/patologia , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Proteínas Repressoras/metabolismo , Proteínas Repressoras/genética , Glucose/metabolismo , Masculino , Transdução de Sinais , Produtos Finais de Glicação Avançada/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/complicações , Encefalopatias/metabolismo , Encefalopatias/etiologia , Encefalopatias/patologia , Complicações do Diabetes/metabolismo , Complicações do Diabetes/patologia , Linhagem Celular , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/etiologia , Mitocôndrias/metabolismo , Camundongos Endogâmicos C57BL
4.
Int J Mol Sci ; 25(20)2024 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-39456683

RESUMO

Brain development is an extremely complex and essential biological process that begins at the start of life and continues throughout an individual's lifespan [...].


Assuntos
Encéfalo , Humanos , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Encéfalo/patologia , Animais , Encefalopatias/metabolismo , Encefalopatias/patologia , Encefalopatias/etiologia , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/patologia
5.
Int J Mol Sci ; 25(20)2024 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-39456734

RESUMO

Microglia signatures refer to distinct gene expression profiles or patterns of gene activity that are characteristic of microglia. Advances in gene expression profiling techniques, such as single-cell RNA sequencing, have allowed us to study microglia at a more detailed level and identify unique gene expression patterns that are associated, but not always, with different functional states of these cells. Microglial signatures depend on the developmental stage, brain region, and specific pathological conditions. By studying these signatures, it has been possible to gain insights into the underlying mechanisms of microglial activation and begin to develop targeted therapies to modulate microglia-mediated immune responses in the CNS. Historically, the first two signatures coincide with M1 pro-inflammatory and M2 anti-inflammatory phenotypes. The first one includes upregulation of genes such as CD86, TNF-α, IL-1ß, and iNOS, while the second one may involve genes like CD206, Arg1, Chil3, and TGF-ß. However, it has long been known that many and more specific phenotypes exist between M1 and M2, likely with corresponding signatures. Here, we discuss specific microglial signatures and their association, if any, with neurodegenerative pathologies and other brain disorders.


Assuntos
Encefalopatias , Microglia , Microglia/metabolismo , Microglia/patologia , Humanos , Animais , Encefalopatias/genética , Encefalopatias/patologia , Encefalopatias/metabolismo , Encefalopatias/etiologia , Transcriptoma , Encéfalo/metabolismo , Encéfalo/patologia , Perfilação da Expressão Gênica
6.
Thromb Res ; 243: 109154, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39305718

RESUMO

Neurological diseases (ND), including neurodegenerative diseases (NDD) and psychiatric disorders (PD), present a significant public health challenge, ranking third in Europe for disability and premature death, following cardiovascular diseases and cancers. In 2017, approximately 540 million cases of ND were reported among Europe's 925 million people, with strokes, dementia, and headaches being most prevalent. Nowadays, more and more evidence highlight the hemostasis critical role in cerebral homeostasis and vascular events. Indeed, hemostasis, thrombosis, and brain abnormalities contributing to ND form a complex and poorly understood equilibrium. Alterations in vascular biology, particularly involving the blood-brain barrier, are implicated in ND, especially dementia, and PD. While the roles of key coagulation players such as thrombin and fibrinogen are established, the roles of other hemostasis components are less clear. Moreover, the involvement of these elements in psychiatric disease pathogenesis is virtually unstudied, except in specific pathological models such as antiphospholipid syndrome. Advanced imaging techniques, primarily functional magnetic resonance imaging and its derivatives like diffusion tensor imaging, have been developed to study brain areas affected by ND and to improve our understanding of the pathophysiology of these diseases. This literature review aims to clarify the current understanding of the connections between hemostasis, thrombosis, and neurological diseases, as well as explore potential future diagnostic and therapeutic strategies.


Assuntos
Encefalopatias , Hemostasia , Humanos , Hemostasia/fisiologia , Encefalopatias/metabolismo , Encefalopatias/sangue , Doença Crônica , Trombose/sangue , Trombose/metabolismo
7.
Cells ; 13(17)2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-39273013

RESUMO

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.


Assuntos
Encéfalo , ATPases Vacuolares Próton-Translocadoras , Humanos , ATPases Vacuolares Próton-Translocadoras/metabolismo , ATPases Vacuolares Próton-Translocadoras/genética , Encéfalo/patologia , Encéfalo/metabolismo , Animais , Lisossomos/metabolismo , Lisossomos/enzimologia , Encefalopatias/genética , Encefalopatias/metabolismo , Encefalopatias/enzimologia , Encefalopatias/patologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo
8.
Neurobiol Dis ; 200: 106641, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39159894

RESUMO

STriatal-Enriched protein tyrosine Phosphatase (STEP) is a brain-specific tyrosine phosphatase that is associated with numerous neurological and neuropsychiatric disorders. STEP dephosphorylates and inactivates various kinases and phosphatases critical for neuronal function and health including Fyn, Pyk2, ERK1/2, p38, and PTPα. Importantly, STEP dephosphorylates NMDA and AMPA receptors, two major glutamate receptors that mediate fast excitatory synaptic transmission. This STEP-mediated dephosphorylation leads to their internalization and inhibits both Hebbian synaptic potentiation and homeostatic synaptic scaling. Hence, STEP has been widely accepted to weaken excitatory synaptic strength. However, emerging evidence implicates a novel role of STEP in neuronal hyperexcitability and seizure disorders. Genetic deletion and pharmacological blockade of STEP reduces seizure susceptibility in acute seizure mouse models and audiogenic seizures in a mouse model of Fragile X syndrome. Pharmacologic inhibition of STEP also decreases hippocampal activity and neuronal intrinsic excitability. Here, we will highlight the divergent roles of STEP in excitatory synaptic transmission and neuronal intrinsic excitability, present the potential underlying mechanisms, and discuss their impact on STEP-associated neurologic and neuropsychiatric disorders.


Assuntos
Proteínas Tirosina Fosfatases não Receptoras , Animais , Humanos , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/genética , Encefalopatias/metabolismo , Encefalopatias/fisiopatologia , Transmissão Sináptica/fisiologia , Convulsões/metabolismo , Convulsões/fisiopatologia , Neurônios/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Fosfatases/genética
9.
Ageing Res Rev ; 100: 102450, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39134179

RESUMO

Diabetic encephalopathy (DE), a significant micro-complication of diabetes, manifests as neurochemical, structural, behavioral, and cognitive alterations. This condition is especially dangerous for the elderly because aging raises the risk of neurodegenerative disorders and cognitive impairment, both of which can be made worse by diabetes. Despite its severity, diagnosis of this disease is challenging, and there is a paucity of information on its pathogenesis. The pivotal roles of various cellular pathways, activated or influenced by hyperglycemia, insulin sensitivity, amyloid accumulation, tau hyperphosphorylation, brain vasculopathy, neuroinflammation, and oxidative stress, are widely recognized for contributing to the potential causes of diabetic encephalopathy. We also reviewed current pharmacological strategies for DE encompassing a comprehensive approach targeting metabolic dysregulations and neurological manifestations. Antioxidant-based therapies hold promise in mitigating oxidative stress-induced neuronal damage, while anti-diabetic drugs offer neuroprotective effects through diverse mechanisms, including modulation of insulin signaling pathways and neuroinflammation. Additionally, tissue engineering and nanomedicine-based approaches present innovative strategies for targeted drug delivery and regenerative therapies for DE. Despite significant progress, challenges remain in translating these therapeutic interventions into clinical practice, including long-term safety, scalability, and regulatory approval. Further research is warranted to optimize these approaches and address remaining gaps in the management of DE and associated neurodegenerative disorders.


Assuntos
Hipoglicemiantes , Humanos , Animais , Hipoglicemiantes/uso terapêutico , Complicações do Diabetes/terapia , Complicações do Diabetes/metabolismo , Estresse Oxidativo/fisiologia , Encefalopatias/terapia , Encefalopatias/etiologia , Encefalopatias/metabolismo
11.
BMC Res Notes ; 17(1): 204, 2024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-39049055

RESUMO

OBJECTIVE: In 2004, after consuming angel-wing mushrooms, Pleurocybella porrigens, 59 incidents of food poisoning were reported in Japan. Consequently, 17 individuals died of acute encephalopathy. In 2023, we proved that a lectin, pleurocybelline, and pleurocybellaziridine from this mushroom caused damage to the brains of mice. Although we reported genomic and transcriptomic data of P. porrigens in 2013, the assembly quality of the transcriptomic data was inadequate for accurate functional annotation. Thus, we obtained detailed transcriptomic data on the fruiting bodies and mycelia of this mushroom using Illumina NovaSeq 6000. RESULTS: De novo assembly data indicated that the N50 lengths for the fruiting bodies and mycelia were improved compared with those previously reported. The differential expression analysis between the fruiting bodies and the mycelia revealed that 1,937 and 1,555 genes were significantly up-regulated in the fruiting bodies and the mycelia, respectively. The biological functions of P. porrigens transcripts, including PA biosynthetic pathways, were investigated using BLAST search, Gene Ontology, and Kyoto Encyclopedia of Genes and Genomes pathway analysis. The obtained results revealed L-valine, a predicted precursor of PA, is biosynthesized in the fruiting bodies and mycelia. Furthermore, real-time RT-PCR was performed to evaluate the accuracy of the results of differential expression analysis.


Assuntos
Carpóforos , Micélio , Carpóforos/genética , Micélio/genética , Camundongos , Animais , Agaricales/genética , Agaricales/metabolismo , RNA-Seq/métodos , Encefalopatias/genética , Encefalopatias/metabolismo , Transcriptoma/genética , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Intoxicação Alimentar por Cogumelos
12.
Diabetes ; 73(9): 1513-1526, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38869375

RESUMO

Diabetic encephalopathy (DE) is a severe complication of the central nervous system associated with diabetes. In this study, we investigated the regulatory role of mammalian target of rapamycin (mTOR) on nuclear factor κB (NF-κB) in mice with DE, and the neuroprotective effect and therapeutic mechanisms of luteolin, a natural flavonoid compound with anti-inflammatory, antioxidant, and neuroprotective properties. The results indicated that treatment with luteolin improved the degree of cognitive impairment in mice with DE. It also decreased the levels of phosphorylated mTOR, phosphorylated NF-κB, and histone deacetylase 2 (HDAC2) and increased the expression of brain-derived neurotrophic factor and synaptic-related proteins. Furthermore, protein-protein interaction and the Gene Ontology analysis revealed that luteolin was involved in the regulatory network of HDAC2 expression through the mTOR/NF-κB signaling cascade. Our bioinformatics and molecular docking results indicated that luteolin may also directly target HDAC2, as an HDAC2 inhibitor, to alleviate DE, complementing mTOR/NF-κB signaling inhibition. Analysis of luteolin's target proteins and their interactions suggest an effect on HDAC2 and cognition. In conclusion, HDAC2 and tau hyperphosphorylation are regulated by the mTOR/NF-κB signaling cascade in DE, and luteolin is found to reverse these effects, demonstrating its protective role in DE.


Assuntos
Histona Desacetilase 2 , Luteolina , NF-kappa B , Proteínas tau , Luteolina/farmacologia , Luteolina/uso terapêutico , Animais , Histona Desacetilase 2/metabolismo , NF-kappa B/metabolismo , Camundongos , Proteínas tau/metabolismo , Fosforilação/efeitos dos fármacos , Masculino , Serina-Treonina Quinases TOR/metabolismo , Transdução de Sinais/efeitos dos fármacos , Encefalopatias/metabolismo , Encefalopatias/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Simulação de Acoplamento Molecular , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Complicações do Diabetes/tratamento farmacológico , Complicações do Diabetes/metabolismo , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Camundongos Endogâmicos C57BL , Fator Neurotrófico Derivado do Encéfalo/metabolismo
13.
CNS Neurosci Ther ; 30(6): e14809, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38923822

RESUMO

BACKGROUND: As physical signals, mechanical cues regulate the neural cells in the brain. The mechanosensitive channels (MSCs) perceive the mechanical cues and transduce them by permeating specific ions or molecules across the plasma membrane, and finally trigger a series of intracellular bioelectrical and biochemical signals. Emerging evidence supports that wide-distributed, high-expressed MSCs like Piezo1 play important roles in several neurophysiological processes and neurological disorders. AIMS: To systematically conclude the functions of MSCs in the brain and provide a novel mechanobiological perspective for brain diseases. METHOD: We summarized the mechanical cues and MSCs detected in the brain and the research progress on the functional roles of MSCs in physiological conditions. We then concluded the pathological activation and downstream pathways triggered by MSCs in two categories of brain diseases, neurodegenerative diseases and place-occupying damages. Finally, we outlined the methods for manipulating MSCs and discussed their medical potential with some crucial outstanding issues. RESULTS: The MSCs present underlying common mechanisms in different brain diseases by acting as the "transportation hubs" to transduce the distinct signal patterns: the upstream mechanical cues and the downstream intracellular pathways. Manipulating the MSCs is feasible to alter the complicated downstream processes, providing them promising targets for clinical treatment. CONCLUSIONS: Recent research on MSCs provides a novel insight into brain diseases. The common mechanisms mediated by MSCs inspire a wide range of therapeutic potentials targeted on MSCs in different brain diseases.


Assuntos
Encefalopatias , Canais Iônicos , Mecanotransdução Celular , Humanos , Animais , Canais Iônicos/metabolismo , Canais Iônicos/fisiologia , Encefalopatias/metabolismo , Encefalopatias/fisiopatologia , Mecanotransdução Celular/fisiologia , Encéfalo/metabolismo
14.
Cell Rep Med ; 5(6): 101609, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38897176

RESUMO

ATP-binding cassette (ABC) transporters facilitate the movement of diverse molecules across cellular membranes, including those within the CNS. While most extensively studied in microvascular endothelial cells forming the blood-brain barrier (BBB), other CNS cell types also express these transporters. Importantly, disruptions in the CNS microenvironment during disease can alter transporter expression and function. Through this comprehensive review, we explore the modulation of ABC transporters in various brain pathologies and the context-dependent consequences of these changes. For instance, downregulation of ABCB1 may exacerbate amyloid beta plaque deposition in Alzheimer's disease and facilitate neurotoxic compound entry in Parkinson's disease. Upregulation may worsen neuroinflammation by aiding chemokine-mediated CD8 T cell influx into multiple sclerosis lesions. Overall, ABC transporters at the BBB hinder drug entry, presenting challenges for effective pharmacotherapy. Understanding the context-dependent changes in ABC transporter expression and function is crucial for elucidating the etiology and developing treatments for brain diseases.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Barreira Hematoencefálica , Encéfalo , Humanos , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Encefalopatias/metabolismo , Encefalopatias/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia
15.
Curr Opin Neurobiol ; 87: 102886, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38901329

RESUMO

The integrated stress response (ISR) is a highly conserved biochemical pathway that regulates protein synthesis. The ISR is activated in response to diverse stressors to restore cellular homeostasis. As such, the ISR is implicated in a wide range of diseases, including brain disorders. However, in the brain, the ISR also has potent influence on processes beyond proteostasis, namely synaptic plasticity, learning and memory. Thus, in the setting of brain diseases, ISR activity may have dual effects on proteostasis and synaptic function. In this review, we consider the ISR's contribution to brain disorders through the lens of its potential effects on synaptic plasticity. From these examples, we illustrate that at times ISR activity may be a "double-edged sword". We also highlight its potential as a therapeutic target to improve circuit function in brain diseases independent of its role in disease pathogenesis.


Assuntos
Encefalopatias , Plasticidade Neuronal , Proteostase , Sinapses , Humanos , Proteostase/fisiologia , Animais , Sinapses/fisiologia , Sinapses/metabolismo , Sinapses/patologia , Encefalopatias/metabolismo , Encefalopatias/fisiopatologia , Plasticidade Neuronal/fisiologia , Estresse Fisiológico/fisiologia , Encéfalo/metabolismo
16.
J Clin Invest ; 134(15)2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38874642

RESUMO

GNAO1 mutated in pediatric encephalopathies encodes the major neuronal G protein Gαo. Of the more than 80 pathogenic mutations, most are single amino acid substitutions spreading across the Gαo sequence. We performed extensive characterization of Gαo mutants, showing abnormal GTP uptake and hydrolysis and deficiencies in binding Gßγ and RGS19. Plasma membrane localization of Gαo was decreased for a subset of mutations that leads to epilepsy; dominant interactions with GPCRs also emerged for the more severe mutants. Pathogenic mutants massively gained interaction with Ric8A and, surprisingly, Ric8B proteins, relocalizing them from cytoplasm to Golgi. Of these 2 mandatory Gα-subunit chaperones, Ric8A is normally responsible for the Gαi/Gαo, Gαq, and Gα12/Gα13 subfamilies, and Ric8B solely responsible for Gαs/Gαolf. Ric8 mediates the disease dominance when engaging in neomorphic interactions with pathogenic Gαo through imbalance of the neuronal G protein signaling networks. As the strength of Gαo-Ric8B interactions correlates with disease severity, our study further identifies an efficient biomarker and predictor for clinical manifestations in GNAO1 encephalopathies. Our work uncovers the neomorphic molecular mechanism of mutations underlying pediatric encephalopathies and offers insights into other maladies caused by G protein malfunctioning and further genetic diseases.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP , Animais , Feminino , Humanos , Masculino , Encefalopatias/genética , Encefalopatias/metabolismo , Encefalopatias/patologia , Drosophila melanogaster , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células HEK293 , Mutação
17.
Neuroscience ; 552: 100-111, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-38936457

RESUMO

Lactate in the brain is produced endogenously and exogenously. The primary functional cells that produce lactate in the brain are astrocytes. Astrocytes release lactate to act on neurons, thereby affecting neuronal function, through a process known as the astrocyte-neuron shuttle. Lactate affects microglial function as well and inhibits microglia-mediated neuroinflammation. Lactate also provides energy, acts as a signaling molecule, and promotes neurogenesis. This article summarizes the role of lactate in cells, animals, and humans. Lactate is a protective molecule against stress in healthy organisms and in the early stages of brain disorders. Thus, lactate may be a potential therapeutic target for brain disorders. Further research on the role of lactate in microglia may have great prospects. This article provides a new perspective and research direction for the study of lacate in brain disorders.


Assuntos
Encefalopatias , Ácido Láctico , Humanos , Animais , Ácido Láctico/metabolismo , Encefalopatias/metabolismo , Encefalopatias/tratamento farmacológico , Astrócitos/metabolismo , Microglia/metabolismo , Encéfalo/metabolismo , Neurônios/metabolismo
18.
Cell Biochem Biophys ; 82(2): 521-534, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38727784

RESUMO

In normal and pathophysiological conditions our cells secrete vesicular bodies known as extracellular particles. Extracellular vesicles are lipid-bound extracellular particles. A majority of these extracellular vesicles are linked to cell-to-cell communication. Brain consists of tightly packed neural cells. Neural cell releases extracellular vesicles in cerebrospinal fluid. Extracellular vesicle mediated crosstalk maintains neural homeostasis in the central nervous system via transferring cargos between neural cells. In neurodegenerative diseases, small extracellular vesicle transfer misfolded proteins to healthy cells in the neural microenvironment. They can also cross blood-brain barrier (BBB) and stimulate peripheral immune response inside central nervous system. In today's world different approaches employ extracellular vesicle in various therapeutics. This review gives a brief knowledge about the biological relevance of extracellular vesicles in the central nervous system and relevant advances in the translational application of EV in brain disorders.


Assuntos
Encéfalo , Vesículas Extracelulares , Humanos , Vesículas Extracelulares/metabolismo , Encéfalo/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Comunicação Celular , Encefalopatias/metabolismo
19.
J Mol Neurosci ; 74(2): 54, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38760510

RESUMO

This article discusses a rare case of coexistent meningiomas and Primary familial brain calcification (PFBC). PFBC is a neurodegenerative disease characterized by brain calcifications and a variety of neuropsychiatric symptoms and signs, with pathogenic variants in specific genes. The study explores the potential link between PFBC and meningiomas, highlighting shared features like intralesional calcifications and common genes such as MEA6. The article also revisits PFBC patients developing other brain tumors, particularly gliomas, emphasizing the intersection of oncogenes like PDGFB and PDGFRB in both calcifications and tumor progression. In recent investigations, attention has extended beyond brain tumors to breast cancer metastasis, unveiling a noteworthy connection. These findings suggest a broader connection between brain calcifications and tumors, encouraging a reevaluation of therapeutic approaches for PFBC.


Assuntos
Neoplasias Encefálicas , Calcinose , Meningioma , Humanos , Calcinose/genética , Calcinose/patologia , Meningioma/genética , Meningioma/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Feminino , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patologia , Encefalopatias/genética , Encefalopatias/patologia , Encefalopatias/metabolismo
20.
Neurosci Biobehav Rev ; 162: 105731, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38763180

RESUMO

Fragile X messenger ribonucleoprotein 1 (FMRP) is a widely expressed RNA binding protein involved in several steps of mRNA metabolism. Mutations in the FMR1 gene encoding FMRP are responsible for fragile X syndrome (FXS), a leading genetic cause of intellectual disability and autism spectrum disorder, and fragile X-associated tremor-ataxia syndrome (FXTAS), a neurodegenerative disorder in aging men. Although FMRP is mainly expressed in neurons, it is also present in glial cells and its deficiency or altered expression can affect functions of glial cells with implications for the pathophysiology of brain disorders. The present review focuses on recent advances on the role of glial subtypes, astrocytes, oligodendrocytes and microglia, in the pathophysiology of FXS and FXTAS, and describes how the absence or reduced expression of FMRP in these cells can impact on glial and neuronal functions. We will also briefly address the role of FMRP in radial glial cells and its effects on neural development, and gliomas and will speculate on the role of glial FMRP in other brain disorders.


Assuntos
Proteína do X Frágil da Deficiência Intelectual , Síndrome do Cromossomo X Frágil , Neuroglia , Humanos , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Neuroglia/metabolismo , Animais , Síndrome do Cromossomo X Frágil/metabolismo , Síndrome do Cromossomo X Frágil/fisiopatologia , Síndrome do Cromossomo X Frágil/patologia , Encefalopatias/metabolismo , Encefalopatias/fisiopatologia , Encefalopatias/genética , Ataxia/metabolismo , Ataxia/fisiopatologia , Ataxia/genética , Tremor/metabolismo , Tremor/fisiopatologia , Tremor/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA