Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 765
Filtrar
1.
Medicina (Kaunas) ; 60(9)2024 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-39336425

RESUMO

Nanomedicine is a newer, promising approach to promote neuroprotection, neuroregeneration, and modulation of the blood-brain barrier. This review includes the integration of various nanomaterials in neurological disorders. In addition, gelatin-based hydrogels, which have huge potential due to biocompatibility, maintenance of porosity, and enhanced neural process outgrowth, are reviewed. Chemical modification of these hydrogels, especially with guanidine moieties, has shown improved neuron viability and underscores tailored biomaterial design in neural applications. This review further discusses strategies to modulate the blood-brain barrier-a factor critically associated with the effective delivery of drugs to the central nervous system. These advances bring supportive solutions to the solving of neurological conditions and innovative therapies for their treatment. Nanomedicine, as applied to neuroscience, presents a significant leap forward in new therapeutic strategies that might help raise the treatment and management of neurological disorders to much better levels. Our aim was to summarize the current state-of-knowledge in this field.


Assuntos
Barreira Hematoencefálica , Nanomedicina , Neuroproteção , Humanos , Nanomedicina/métodos , Nanomedicina/tendências , Neuroproteção/fisiologia , Regeneração Nervosa/efeitos dos fármacos , Regeneração Nervosa/fisiologia , Fármacos Neuroprotetores/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/terapia , Hidrogéis/uso terapêutico
2.
Eur J Neurosci ; 60(8): 6046-6056, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39297873

RESUMO

Menopause weakens the brain's structural integrity and increases its susceptibility to a range of degenerative and mental illnesses. 17ß estradiol (17ßE2) exhibits potent neuroprotective properties. Exogenous estrogen supplementation provides neuroprotection, but the findings presented by the Million Women Study (MWS) and the Women's Health Initiative (WHI), as well as the increased risk of endometrial cancer, breast cancer and venous thromboembolism associated with estrogen use, have cast doubt on its clinical use for neurological disorders. Thus, the objective of our review article is to compile all in vitro and in vivo studies conducted till date demonstrating the neuroprotective potential of nonfeminizing estrogens. This objective has been achieved by gathering various research and review manuscripts from different records such as PubMed, Embase, Scopus, Google Scholar, Web of Science and OVID, using different terms like 'estrogen deficiency, 17ß estradiol, non-feminising estrogens, and brain disorder'. However, recent evidence has revealed the contribution of numerous non-estrogen receptor-dependent pathways in neuroprotective effects of estrogen. In conclusion, synthetic nonfeminizing estrogens that have little or no ER binding but are equally powerful (and in some cases more potent) in delivering neuroprotection are emerging as viable and potential alternatives.


Assuntos
Estrogênios , Fármacos Neuroprotetores , Humanos , Fármacos Neuroprotetores/farmacologia , Animais , Estrogênios/farmacologia , Estradiol/farmacologia , Neuroproteção/efeitos dos fármacos , Neuroproteção/fisiologia , Feminino
3.
Acta Neuropathol Commun ; 12(1): 150, 2024 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-39300576

RESUMO

BACKGROUND: Glaucoma is a leading cause of blindness, affecting retinal ganglion cells (RGCs) and their axons. By 2040, it is likely to affect 110 million people. Neuroinflammation, specifically through the release of proinflammatory cytokines by M1 microglial cells, plays a crucial role in glaucoma progression. Indeed, in post-mortem human studies, pre-clinical models, and ex-vivo models, RGC degeneration has been consistently shown to be linked to inflammation in response to cell death and tissue damage. Recently, Rho kinase inhibitors (ROCKis) have emerged as potential therapies for neuroinflammatory and neurodegenerative diseases. This study aimed to investigate the potential effects of three ROCKis (Y-27632, Y-33075, and H-1152) on retinal ganglion cell (RGC) loss and retinal neuroinflammation using an ex-vivo retinal explant model. METHODS: Rat retinal explants underwent optic nerve axotomy and were treated with Y-27632, Y-33075, or H-1152. The neuroprotective effects on RGCs were evaluated using immunofluorescence and Brn3a-specific markers. Reactive glia and microglial activation were studied by GFAP, CD68, and Iba1 staining. Flow cytometry was used to quantify day ex-vivo 4 (DEV 4) microglial proliferation and M1 activation by measuring the number of CD11b+, CD68+, and CD11b+/CD68+ cells after treatment with control solvent or Y-33075. The modulation of gene expression was measured by RNA-seq analysis on control and Y-33075-treated explants and glial and pro-inflammatory cytokine gene expression was validated by RT-qPCR. RESULTS: Y-27632 and H-1152 did not significantly protect RGCs. By contrast, at DEV 4, 50 µM Y-33075 significantly increased RGC survival. Immunohistology showed a reduced number of Iba1+/CD68+ cells and limited astrogliosis with Y-33075 treatment. Flow cytometry confirmed lower CD11b+, CD68+, and CD11b+/CD68+ cell numbers in the Y-33075 group. RNA-seq showed Y-33075 inhibited the expression of M1 microglial markers (Tnfα, Il-1ß, Nos2) and glial markers (Gfap, Itgam, Cd68) and to reduce apoptosis, ferroptosis, inflammasome formation, complement activation, TLR pathway activation, and P2rx7 and Gpr84 gene expression. Conversely, Y-33075 upregulated RGC-specific markers, neurofilament formation, and neurotransmitter regulator expression, consistent with its neuroprotective effects. CONCLUSION: Y-33075 demonstrates marked neuroprotective and anti-inflammatory effects, surpassing the other tested ROCKis (Y-27632 and H-1152) in preventing RGC death and reducing microglial inflammatory responses. These findings highlight its potential as a therapeutic option for glaucoma.


Assuntos
Fármacos Neuroprotetores , Piridinas , Células Ganglionares da Retina , Quinases Associadas a rho , Animais , Piridinas/farmacologia , Quinases Associadas a rho/metabolismo , Quinases Associadas a rho/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Ratos , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia , Células Ganglionares da Retina/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Retina/efeitos dos fármacos , Retina/patologia , Retina/metabolismo , Amidas/farmacologia , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Ratos Sprague-Dawley , Neuroproteção/efeitos dos fármacos , Neuroproteção/fisiologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Inibidores de Proteínas Quinases/farmacologia , Masculino , Traumatismos do Nervo Óptico/tratamento farmacológico , Traumatismos do Nervo Óptico/patologia , Traumatismos do Nervo Óptico/metabolismo , Isoquinolinas , Sulfonamidas
4.
Pflugers Arch ; 476(11): 1703-1725, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39136758

RESUMO

Precise control of norepinephrine (NE) levels and NE-receptor interaction is crucial for proper function of the brain. Much evidence for this view comes from experimental studies that indicate an important role for NE in the pathophysiology and treatment of various conditions, including cognitive dysfunction, Alzheimer's disease, Parkinson's disease, multiple sclerosis, and sleep disorders. NE provides neuroprotection against several types of insults in multiple ways. It abrogates oxidative stress, attenuates neuroinflammatory responses in neurons and glial cells, reduces neuronal and glial cell activity, promotes autophagy, and ameliorates apoptotic responses to a variety of insults. It is beneficial for the treatment of neurodegenerative diseases because it improves the generation of neurotrophic factors, promotes neuronal survival, and plays an important role in the regulation of adult neurogenesis. This review aims to present the evidence supporting a principal role for NE in neuroprotection, and molecular mechanisms of neuroprotection.


Assuntos
Fármacos Neuroprotetores , Norepinefrina , Humanos , Animais , Norepinefrina/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/tratamento farmacológico , Neuroproteção/fisiologia , Neuroproteção/efeitos dos fármacos
5.
Inflammopharmacology ; 32(5): 2801-2820, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39136812

RESUMO

Parkinson's disease (PD) is an age-related chronic neurological condition characterized by progressive degeneration of dopaminergic neurons and the presence of Lewy bodies, primarily composed of alpha-synuclein and ubiquitin. The pathophysiology of PD encompasses alpha-synuclein aggregation, oxidative stress, neuroinflammation, mitochondrial dysfunction, and impaired autophagy and ubiquitin-proteasome systems. Among these, the Keap1-Nrf2 pathway is a key regulator of antioxidant defense mechanisms. Nrf2 has emerged as a crucial factor in managing oxidative stress and inflammation, and it also influences ubiquitination through p62 expression. Keap1 negatively regulates Nrf2 by targeting it for degradation via the ubiquitin-proteasome system. Disruption of the Nrf2-Keap1 pathway in PD affects cellular responses to oxidative stress and inflammation, thereby playing a critical role in disease progression. In addition, the role of neuroinflammation in PD has gained significant attention, highlighting the interplay between immune responses and neurodegeneration. This review discusses the various mechanisms responsible for neuronal degeneration in PD, with a special emphasis on the neuroprotective role of the Nrf2-Keap1 pathway. Furthermore, it explores the implications of inflammopharmacology in modulating these pathways to provide therapeutic insights for PD.


Assuntos
Proteína 1 Associada a ECH Semelhante a Kelch , Fator 2 Relacionado a NF-E2 , Neuroproteção , Estresse Oxidativo , Doença de Parkinson , Humanos , Fator 2 Relacionado a NF-E2/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/tratamento farmacológico , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Animais , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Neuroproteção/fisiologia , Transdução de Sinais/fisiologia , Fármacos Neuroprotetores/farmacologia , Inflamação/metabolismo , Inflamação/tratamento farmacológico
6.
Psychother Psychosom ; 93(5): 285-291, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39154647

RESUMO

Neuroprotection aims to safeguard neurons from damage caused by various factors like stress, potentially leading to the rescue, recovery, or regeneration of the nervous system and its functions [J Clin Neurosci. 2002;9(1):4-8]. Conversely, neuroplasticity refers to the brain's ability to adapt and change throughout life, involving structural and functional alterations in cells and synaptic transmission [Neural Plast. 2014;2014:541870]. Neuroprotection is a broad and multidisciplinary field encompassing various approaches and strategies aimed at preserving and promoting neuronal health. It is a critical area of research in neuroscience and neurology, with the potential to lead to new therapies for a wide range of neurological disorders and conditions. Neuroprotection can take various forms and may involve pharmacological agents, lifestyle modifications, or behavioral interventions. Accordingly, also the perspective and the meaning of neuroprotection differs due to different angles of interpretation. The primary interpretation is from the pharmacological point of view since the most consistent data come from this field. In addition, we will discuss also alternative, yet less considered, perspectives on neuroprotection, focusing on specific neuroprotective targets, interactions with surrounding microglia, different levels of neuroprotective effects, the reversive/adaptative dimension, and its use as anticipatory/prophylactic intervention.


Assuntos
Neuroproteção , Fármacos Neuroprotetores , Humanos , Neuroproteção/fisiologia , Fármacos Neuroprotetores/uso terapêutico , Plasticidade Neuronal/fisiologia
7.
Prog Brain Res ; 289: 169-180, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39168579

RESUMO

Coffee is a popular drink enjoyed around the world, and scientists are very interested in studying how it affects the human brain. This chapter looks at lots of different studies to understand how drinking coffee might change the brain and help protect it from neurodegenerative disorders especially like schizophrenia. With the help of available literature a link between the coffee mechanism and neurodegenerative disorders is established in this chapter. Researchers have found that drinking coffee can change the size of certain parts of the brain that control things like thinking and mood. Scientists also study how coffee's ingredients, especially caffeine, can change how the brain works. They think these changes could help protect the brain from diseases. This chapter focuses on how coffee might affect people with schizophrenia as hallucination is caused during and after excess consumption of caffeine. There's still a lot we don't know, but researchers are learning more by studying how different people's brains respond to coffee over time. Overall, this chapter shows that studying coffee and the brain could lead to new ways to help people with brain disorders. This study also draws ideas for future research and ways to help people stay healthy.


Assuntos
Café , Substância Cinzenta , Humanos , Substância Cinzenta/efeitos dos fármacos , Substância Cinzenta/patologia , Neuroproteção/fisiologia , Neuroproteção/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Cafeína/farmacologia , Cafeína/administração & dosagem , Doenças do Sistema Nervoso/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Animais , Esquizofrenia
8.
Acta Neuropathol Commun ; 12(1): 137, 2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39180087

RESUMO

A compromised capacity to maintain NAD pools is recognized as a key underlying pathophysiological feature of neurodegenerative diseases. NAD acts as a substrate in major cell functions including mitochondrial homeostasis, cell signalling, axonal transport, axon/Wallerian degeneration, and neuronal energy supply. Dendritic degeneration is an early marker of neuronal stress and precedes cell loss. However, little is known about dendritic structural preservation in pathologic environments and remodelling in mature neurons. Retinal ganglion cell dendritic atrophy is an early pathological feature in animal models of the disease and has been demonstrated in port-mortem human glaucoma samples. Here we report that a nicotinamide (a precursor to NAD through the NAD salvage pathway) enriched diet provides robust retinal ganglion cell dendritic protection and preserves dendritic structure in a rat model of experimental glaucoma. Metabolomic analysis of optic nerve samples from the same animals demonstrates that nicotinamide provides robust metabolic neuroprotection in glaucoma. Advances in our understanding of retinal ganglion cell metabolic profiles shed light on the energetic shift that triggers early neuronal changes in neurodegenerative diseases. As nicotinamide can improve visual function short term in existing glaucoma patients, we hypothesize that a portion of this visual recovery may be due to dendritic preservation in stressed, but not yet fully degenerated, retinal ganglion cells.


Assuntos
Modelos Animais de Doenças , Glaucoma , Fármacos Neuroprotetores , Niacinamida , Células Ganglionares da Retina , Animais , Niacinamida/farmacologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia , Células Ganglionares da Retina/metabolismo , Glaucoma/metabolismo , Glaucoma/patologia , Fármacos Neuroprotetores/farmacologia , Ratos , Relação Dose-Resposta a Droga , Masculino , Administração Oral , Nervo Óptico/efeitos dos fármacos , Nervo Óptico/patologia , Nervo Óptico/metabolismo , Neuroproteção/efeitos dos fármacos , Neuroproteção/fisiologia , Dendritos/efeitos dos fármacos , Dendritos/patologia , Dendritos/metabolismo , Complexo Vitamínico B/farmacologia , Complexo Vitamínico B/administração & dosagem
9.
J Neuroinflammation ; 21(1): 189, 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39095837

RESUMO

BACKGROUND: Infection with the protozoan parasite Toxoplasma gondii leads to the formation of lifelong cysts in neurons that can have devastating consequences in the immunocompromised. In the immunocompetent individual, anti-parasitic effector mechanisms and a balanced immune response characterized by pro- and anti-inflammatory cytokine production establishes an asymptomatic infection that rarely leads to neurological symptoms. Several mechanisms are known to play a role in this successful immune response in the brain including T cell production of IFNγ and IL-10 and the involvement of CNS resident cells. This limitation of clinical neuropathology during chronic infection suggests a balance between immune response and neuroprotective mechanisms that collectively prevent clinical manifestations of disease. However, how these two vital mechanisms of protection interact during chronic Toxoplasma infection remains poorly understood. MAIN TEXT: This study demonstrates a previously undescribed connection between innate neutrophils found chronically in the brain, termed "chronic brain neutrophils" (CBNeuts), and neuroprotective mechanisms during Toxoplasma infection. Lack of CBNeuts during chronic infection, accomplished via systemic neutrophil depletion, led to enhanced infection and deleterious effects on neuronal regeneration and repair mechanisms in the brain. Phenotypic and transcriptomic analysis of CBNeuts identified them as distinct from peripheral neutrophils and revealed two main subsets of CBNeuts that display heterogeneity towards both classical effector and neuroprotective functions in an age-dependent manner. Further phenotypic profiling defined expression of the neuroprotective molecules NRG-1 andErbB4 by these cells, and the importance of this signaling pathway during chronic infection was demonstrated via NRG-1 treatment studies. CONCLUSIONS: In conclusion, this work identifies CBNeuts as a heterogenous population geared towards both classical immune responses and neuroprotection during chronic Toxoplasma infection and provides the foundation for future mechanistic studies of these cells.


Assuntos
Neutrófilos , Toxoplasmose , Animais , Neutrófilos/imunologia , Neutrófilos/metabolismo , Camundongos , Toxoplasmose/imunologia , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/imunologia , Toxoplasma/imunologia , Feminino , Neuroproteção/fisiologia , Masculino , Encéfalo/imunologia , Encéfalo/patologia , Encéfalo/parasitologia
11.
CNS Neurosci Ther ; 30(8): e14836, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39097918

RESUMO

INTRODUCTION: Cerebral ischemia-reperfusion injury (CIRI) is a common and debilitating complication of cerebrovascular diseases such as stroke, characterized by mitochondrial dysfunction and cell apoptosis. Unraveling the molecular mechanisms behind these processes is essential for developing effective CIRI treatments. This study investigates the role of RACK1 (receptor for activated C kinase 1) in CIRI and its impact on mitochondrial autophagy. METHODS: We utilized high-throughput transcriptome sequencing and weighted gene co-expression network analysis (WGCNA) to identify core genes associated with CIRI. In vitro experiments used human neuroblastoma SK-N-SH cells subjected to oxygen and glucose deprivation (OGD) to simulate ischemia, followed by reperfusion (OGD/R). RACK1 knockout cells were created using CRISPR/Cas9 technology, and cell viability, apoptosis, and mitochondrial function were assessed. In vivo experiments involved middle cerebral artery occlusion/reperfusion (MCAO/R) surgery in rats, evaluating neurological function and cell apoptosis. RESULTS: Our findings revealed that RACK1 expression increases during CIRI and is protective by regulating mitochondrial autophagy through the PINK1/Parkin pathway. In vitro, RACK1 knockout exacerbated cell apoptosis, while overexpression of RACK1 reversed this process, enhancing mitochondrial function. In vivo, RACK1 overexpression reduced cerebral infarct volume and improved neurological deficits. The regulatory role of RACK1 depended on the PINK1/Parkin pathway, with RACK1 knockout inhibiting PINK1 and Parkin expression, while RACK1 overexpression restored them. CONCLUSION: This study demonstrates that RACK1 safeguards against neural damage in CIRI by promoting mitochondrial autophagy through the PINK1/Parkin pathway. These findings offer crucial insights into the regulation of mitochondrial autophagy and cell apoptosis by RACK1, providing a promising foundation for future CIRI treatments.


Assuntos
Autofagia , Mitocôndrias , Proteínas Quinases , Receptores de Quinase C Ativada , Traumatismo por Reperfusão , Ubiquitina-Proteína Ligases , Animais , Humanos , Ratos , Apoptose/fisiologia , Autofagia/fisiologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Linhagem Celular Tumoral , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/metabolismo , Mitocôndrias/metabolismo , Proteínas de Neoplasias , Neuroproteção/fisiologia , Proteínas Quinases/metabolismo , Proteínas Quinases/genética , Ratos Sprague-Dawley , Receptores de Quinase C Ativada/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transdução de Sinais/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética
12.
Exp Neurol ; 380: 114900, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39059736

RESUMO

AIMS: Adipose-derived stem cell (ADSC)-derived exosomes have been recognized for their neuroprotective effects in various neurological diseases. This study investigates the potential neuroprotective effects of ADSC-derived exosomes in sepsis-associated encephalopathy (SAE). METHODS: Behavioral cognitive functions were evaluated using the open field test, Y-maze test, and novel object recognition test. Brain activity was assessed through functional magnetic resonance imaging (fMRI). Pyroptosis was measured using immunofluorescence staining and western blotting. RESULTS: Our findings indicate that ADSC-derived exosomes mitigate cognitive impairment, improve survival rates, and prevent weight loss in SAE mice. Additionally, exosomes protect hippocampal function in SAE mice, as demonstrated by fMRI evaluations. Furthermore, SAE mice exhibit neuronal damage and infiltration of inflammatory cells in the hippocampus, conditions which are reversed by exosome treatment. Moreover, our study highlights the downstream regulatory role of the NLRP3/caspase-1/GSDMD signaling pathway as a crucial mechanism in alleviating hippocampal inflammation. CONCLUSION: ADSC-derived exosomes confer neuroprotection in SAE models by mediating the NLRP3/caspase-1/GSDMD pathway, thereby ameliorating cognitive impairment.


Assuntos
Caspase 1 , Exossomos , Hipocampo , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR , Piroptose , Encefalopatia Associada a Sepse , Animais , Piroptose/fisiologia , Exossomos/metabolismo , Exossomos/transplante , Hipocampo/metabolismo , Hipocampo/patologia , Encefalopatia Associada a Sepse/metabolismo , Camundongos , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Caspase 1/metabolismo , Neuroproteção/fisiologia , Gasderminas , Proteínas de Ligação a Fosfato
13.
Neurochem Res ; 49(10): 2871-2887, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39026086

RESUMO

Brain injury caused by stroke has a high rate of mortality and remains a major medical challenge worldwide. In recent years, there has been significant attention given to the use of human Umbilical cord-derived Mesenchymal Stem Cells (hUC-MSCs) for the treatment of stroke in different adult and neonate animal models of stroke. However, using hUC-MSCs by systemic administration to treat ischemic stroke has not been investigated sufficiently. In this study, we conducted various experiments to explore the neuroprotection of hUC-MSCs in rats. Our findings demonstrate that an intravenous injection of a high dose of hUC-MSCs at 2 × 10^7 cells/kg markedly ameliorated brain injury resulting from ischemic stroke. This improvement was observed one day after inducing transient middle cerebral artery occlusion (MCAO) and subsequent reperfusion in rats. Notably, the efficacy of this single administration of hUC-MSCs surpassed that of edaravone, even when the latter was used continuously over three days. Mechanistically, secretory factors derived from hUC-MSCs, such as HGF, BDNF, and TNFR1, ameliorated the levels of MDA and T-SOD to regulate oxidative stress. In particular, TNFR1 also improved the expression of NQO-1 and HO-1, important proteins associated with oxidative stress. More importantly, TNFR1 played a significant role in reducing inflammation by modulating IL-6 levels in the blood. Furthermore, TNFR1 was observed to influence the permeability of the blood-brain barrier (BBB) as demonstrated in the evan's blue experiment and protein expression of ZO-1. This study represented a breakthrough in traditional methods and provided a novel strategy for clinical medication and trials.


Assuntos
AVC Isquêmico , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Estresse Oxidativo , Ratos Sprague-Dawley , Cordão Umbilical , Animais , Estresse Oxidativo/fisiologia , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Cordão Umbilical/citologia , Masculino , AVC Isquêmico/metabolismo , AVC Isquêmico/terapia , Ratos , Inflamação/metabolismo , Lesões Encefálicas/metabolismo , Lesões Encefálicas/terapia , Neuroproteção/fisiologia , Infarto da Artéria Cerebral Média/terapia , Infarto da Artéria Cerebral Média/metabolismo
15.
Am J Physiol Regul Integr Comp Physiol ; 327(3): R304-R318, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38860282

RESUMO

Clinical trials of hypothermia after pediatric cardiac arrest (CA) have not seen robust improvement in functional outcome, possibly because of the long delay in achieving target temperature. Previous work in infant piglets showed that high nasal airflow, which induces evaporative cooling in the nasal mucosa, reduced regional brain temperature uniformly in half the time needed to reduce body temperature. Here, we evaluated whether initiation of hypothermia with high transnasal airflow provides neuroprotection without adverse effects in the setting of asphyxic CA. Anesthetized piglets underwent sham-operated procedures (n = 7) or asphyxic CA with normothermic recovery (38.5°C; n = 9) or hypothermia initiated by surface cooling at 10 (n = 8) or 120 (n = 7) min or transnasal cooling initiated at 10 (n = 7) or 120 (n = 7) min after resuscitation. Hypothermia was sustained at 34°C with surface cooling until 20 h followed by 6 h of rewarming. At 4 days of recovery, significant neuronal loss occurred in putamen and sensorimotor cortex. Transnasal cooling initiated at 10 min significantly rescued the number of viable neurons in putamen, whereas levels in putamen in other hypothermic groups remained less than sham levels. In sensorimotor cortex, neuronal viability in the four hypothermic groups was not significantly different from the sham group. These results demonstrate that early initiation of high transnasal airflow in a pediatric CA model is effective in protecting vulnerable brain regions. Because of its simplicity, portability, and low cost, transnasal cooling potentially could be deployed in the field or emergency room for early initiation of brain cooling after pediatric CA.NEW & NOTEWORTHY The onset of therapeutic hypothermia after cardiac resuscitation is often delayed, leading to incomplete neuroprotection. In an infant swine model of asphyxic cardiac arrest, initiation of high transnasal airflow to maximize nasal evaporative cooling produced hypothermia sufficient to provide neuroprotection that was not inferior to body surface cooling. Because of its simplicity and portability, this technique may be of use in the field or emergency room for rapid brain cooling in pediatric cardiac arrest victims.


Assuntos
Modelos Animais de Doenças , Parada Cardíaca , Hipotermia Induzida , Animais , Hipotermia Induzida/métodos , Parada Cardíaca/terapia , Parada Cardíaca/fisiopatologia , Suínos , Neuroproteção/fisiologia , Animais Recém-Nascidos , Feminino , Masculino
16.
Neurochem Res ; 49(9): 2535-2555, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38888830

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the accumulation of amyloid-ß, leading to N-methyl-D-aspartate (NMDA) receptor-dependent synaptic depression, spine elimination, and memory deficits. Glycine transporter type 1 (GlyT1) modulates glutamatergic neurotransmission via NMDA receptors (NMDAR), presenting a potential alternative therapeutic approach for AD. This study investigates the neuroprotective potential of GlyT1 inhibition in an amyloid-ß-induced AD mouse model. C57BL/6 mice were treated with N-[3-([1,1-Biphenyl]-4-yloxy)-3-(4-fluorophenyl)propyl]-N-methylglycine (NFPS), a GlyT1 inhibitor, 24 h prior to intrahippocampal injection of amyloid-ß. NFPS pretreatment prevented amyloid-ß-induced cognitive deficits in short-term and long-term memory, evidenced by novel object recognition and spatial memory tasks. Moreover, NFPS pretreatment curbed microglial activation, astrocytic reactivity, and subsequent neuronal damage from amyloid-ß injection. An extensive label-free quantitative UPLC-MSE proteomic analysis was performed on the hippocampi of mice treated with NFPS. In proteomics, KEGG enrichment analysis revealed increased in dopaminergic synapse, purine-containing compound biosynthetic process and long-term potentiation, and a reduction in Glucose catabolic process and glycolytic process pathways. The western blot analysis confirmed that NFPS treatment elevated BDNF levels, correlating with enhanced TRKB phosphorylation and mTOR activation. Moreover, NFPS treatment reduced the GluN2B expression after 6 h, which was associated with an increase on CaMKIV and CREB phosphorylation. Collectively, these findings demonstrate that GlyT1 inhibition by NFPS activates diverse neuroprotective pathways, enhancing long-term potentiation signaling and countering amyloid-ß-induced hippocampal damage.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Proteínas da Membrana Plasmática de Transporte de Glicina , Hipocampo , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores , Animais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/metabolismo , Masculino , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Camundongos , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de Glicina/antagonistas & inibidores , Proteínas da Membrana Plasmática de Transporte de Glicina/metabolismo , Modelos Animais de Doenças , Sarcosina/análogos & derivados , Sarcosina/farmacologia , Sarcosina/uso terapêutico , Neuroproteção/efeitos dos fármacos , Neuroproteção/fisiologia
17.
Eur J Pediatr ; 183(9): 3647-3653, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38858228

RESUMO

Neuromonitoring has been widely accepted as an important part in neonatal care. Amplitude-integrated EEG (aEEG) and near-infrared spectroscopy (NIRS) are often mentioned in this context, though being only a part of the fully array of methods and examinations that could be considered neuromonitoring. Within the broad array of medical conditions that could be encountered in a neonatal patient, it is important to be aware of the indications for neuromonitoring and especially which neuromonitoring technique to use best for the individual condition. aEEG is now a widely accepted neuromonitor in neonatology with its value in hypoxic events and seizures only rarely questioned. Other methods like NIRS still have to prove themselves in the future. The SafeBoosC-III trial showed that it still remains difficult for some of these methods to prove their value for the improvement of outcome. Bute future developments such as multimodal neuromonitoring with data integration and artificial intelligence analysis could improve the value of these methods.


Assuntos
Eletroencefalografia , Unidades de Terapia Intensiva Neonatal , Espectroscopia de Luz Próxima ao Infravermelho , Humanos , Recém-Nascido , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Eletroencefalografia/métodos , Monitorização Neurofisiológica/métodos , Neuroproteção/fisiologia , Hipóxia-Isquemia Encefálica/diagnóstico , Hipóxia-Isquemia Encefálica/prevenção & controle , Hipóxia-Isquemia Encefálica/terapia , Convulsões/diagnóstico , Convulsões/prevenção & controle , Terapia Intensiva Neonatal/métodos
18.
Brain Res ; 1842: 149104, 2024 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-38945469

RESUMO

Prolactin has been recognized as neuroprotective hormone against various types of neuronal damage. This study was aimed to determine if prolactin protects against streptozotocin injury. A series of experiments were performed to determine neuronal survival by counting total neurons in medial hippocampus cortex and cerebellum. Astrogliosis was determined by immunofluorescence assays using GFAP, and behavioral improvement by prolactin after neuronal damage was determined by open-field and light-dark box tests. Results demonstrated that prolactin induced significant neuronal survival in both the hippocampus and cortex, but not in the cerebellum. No increase in astrogliosis was identified, but a significant reduction in anxiety levels was observed. Overall data indicate that prolactin may protect against a complex form of cell damage including oxidant stress and metabolic disruption by streptozotocin. Prolactin may be helpful strategy in the treatment of neuronal damage in neurological diseases.


Assuntos
Hipocampo , Neurônios , Fármacos Neuroprotetores , Prolactina , Estreptozocina , Animais , Prolactina/metabolismo , Masculino , Fármacos Neuroprotetores/farmacologia , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Ratos , Neuroproteção/fisiologia , Neuroproteção/efeitos dos fármacos , Ratos Sprague-Dawley , Gliose/metabolismo , Cerebelo/metabolismo , Cerebelo/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia
19.
Neurochem Res ; 49(8): 2249-2270, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38837092

RESUMO

Morphine (Mor) has exhibited efficacy in safeguarding neurons against ischemic injuries by simulating ischemic/hypoxic preconditioning (I/HPC). Concurrently, autophagy plays a pivotal role in neuronal survival during IPC against ischemic stroke. However, the involvement of autophagy in Mor-induced neuroprotection and the potential mechanisms remain elusive. Our experiments further confirmed the effect of Mor in cellular and animal models of ischemic stroke and explored its potential mechanism. The findings revealed that Mor enhanced cell viability in a dose-dependent manner by augmenting autophagy levels and autophagic flux in neurons subjected to oxygen-glucose deprivation/reoxygenation (OGD/R). Pretreatment of Mor improved neurological outcome and reduced infarct size in mice with middle cerebral artery occlusion/reperfusion (MCAO/R) at 1, 7 and 14 days. Moreover, the use of autophagy inhibitors nullified the protective effects of Mor, leading to reactive oxygen species (ROS) accumulation, increased loss of mitochondrial membrane potential (MMP) and neuronal apoptosis in OGD/R neurons. Results further demonstrated that Mor-induced autophagy activation was regulated by mTOR-independent activation of the c-Jun NH2- terminal kinase (JNK)1/2 Pathway, both in vitro and in vivo. Overall, these findings suggested Mor-induced neuroprotection by activating autophagy, which were regulated by JNK1/2 pathway in ischemic stroke.


Assuntos
Autofagia , AVC Isquêmico , Morfina , Fármacos Neuroprotetores , Serina-Treonina Quinases TOR , Animais , Autofagia/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Masculino , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/metabolismo , AVC Isquêmico/patologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Morfina/farmacologia , Morfina/uso terapêutico , Camundongos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos Endogâmicos C57BL , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Neuroproteção/efeitos dos fármacos , Neuroproteção/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Espécies Reativas de Oxigênio/metabolismo
20.
CNS Neurosci Ther ; 30(6): e14795, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38867401

RESUMO

AIM: Recent studies have extensively investigated hypothermia as a therapeutic approach for mitigating neural damage. Despite this, bibliometric analyses specifically focusing on this area remain scarce. Consequently, this study aims to comprehensively outline the historical framework of research and to pinpoint future research directions and trends. METHODS: Articles spanning from 2003 to 2023, relevant to both "neuroprotection" and "hypothermia", were sourced from the Web of Science Core Collection. The CiteSpace software facilitated a comprehensive evaluation and analysis of these publications. This analysis included examining the annual productivity, collaboration among nations, institutions, and authors, as well as the network of co-cited references, authors and journals, and the co-occurrence of keywords, and their respective clusters and trends, all of which were visualized. RESULTS: This study included 2103 articles on the neuroprotection effects of hypothermia, noting a consistent increase in publications since 1992. The United States, the University of California System, and Ji Xunming emerged as the most productive nation, institution, and author, respectively. Analysis of the top 10 co-cited publications revealed that seven articles focused on the effects of hypothermia in infants with hypoxic-ischemic encephalopathy, while three studies addressed cardiac arrest. Shankaran S and the journal Stroke were the most frequently co-cited author and journal, respectively. Keyword cluster analysis identified ischemic stroke as the primary focus of hypothermia therapy historically, with cardiac arrest and neonatal hypoxic-ischemic encephalopathy emerging as current research foci. CONCLUSIONS: Recent studies on the neuroprotective effects of hypothermia in cardiac arrest and neonatal hypoxic-ischemic encephalopathy suggest that hypothermia may mitigate neural damage associated with these conditions. However, the application of hypothermia in the treatment of ischemic stroke remains confined to animal models and in vitro studies, with a notable absence of evidence from multicenter randomized controlled trials (RCTs). Further research is required to address this gap.


Assuntos
Bibliometria , Hipotermia Induzida , Neuroproteção , Hipotermia Induzida/tendências , Hipotermia Induzida/métodos , Humanos , Neuroproteção/fisiologia , Animais , Hipóxia-Isquemia Encefálica/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA