Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33.289
Filtrar
1.
CNS Neurosci Ther ; 30(7): e14825, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38954749

RESUMO

AIMS: Ischemic stroke remains a challenge in medical research because of the limited treatment options. Recombinant human tissue plasminogen activator (rtPA) is the primary treatment for recanalization. However, nearly 50% of the patients experience complications that result in ineffective reperfusion. The precise factors contributing to ineffective reperfusion remain unclear; however, recent studies have suggested that immune cells, notably neutrophils, may influence the outcome of rtPA thrombolysis via mechanisms such as the formation of neutrophil extracellular traps. This study aimed to explore the nonthrombolytic effects of rtPA on neutrophils and highlight their contribution to ineffective reperfusion. METHODS: We evaluated the effects of rtPA treatment on middle cerebral artery occlusion in rats. We also assessed neutrophil infiltration and activation after rtPA treatment in vitro and in vivo in a small cohort of patients with massive cerebral ischemia (MCI). RESULTS: rtPA increased neutrophil infiltration into the brain microvessels and worsened blood-brain barrier damage during ischemia. It also increased the neutrophil counts of the patients with MCI. CONCLUSION: Neutrophils play a crucial role in promoting ischemic injury and blood-brain barrier disruption, making them potential therapeutic targets.


Assuntos
Fibrinolíticos , Neutrófilos , Proteínas Recombinantes , Ativador de Plasminogênio Tecidual , Ativador de Plasminogênio Tecidual/farmacologia , Ativador de Plasminogênio Tecidual/uso terapêutico , Animais , Humanos , Masculino , Neutrófilos/efeitos dos fármacos , Ratos , Proteínas Recombinantes/farmacologia , Fibrinolíticos/farmacologia , Fibrinolíticos/uso terapêutico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Ratos Sprague-Dawley , Idoso , Barreira Hematoencefálica/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Feminino , Infiltração de Neutrófilos/efeitos dos fármacos , Pessoa de Meia-Idade , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/imunologia , Modelos Animais de Doenças
2.
Proc Natl Acad Sci U S A ; 121(28): e2322577121, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38968104

RESUMO

Multiple sclerosis (MS) is a demyelinating central nervous system (CNS) disorder that is associated with functional impairment and accruing disability. There are multiple U.S. Food and Drug Administration (FDA)-approved drugs that effectively dampen inflammation and slow disability progression. However, these agents do not work well for all patients and are associated with side effects that may limit their use. The vagus nerve (VN) provides a direct communication conduit between the CNS and the periphery, and modulation of the inflammatory reflex via electrical stimulation of the VN (VNS) shows efficacy in ameliorating pathology in several CNS and autoimmune disorders. We therefore investigated the impact of VNS in a rat experimental autoimmune encephalomyelitis (EAE) model of MS. In this study, VNS-mediated neuroimmune modulation is demonstrated to effectively decrease EAE disease severity and duration, infiltration of neutrophils and pathogenic lymphocytes, myelin damage, blood-brain barrier disruption, fibrinogen deposition, and proinflammatory microglial activation. VNS modulates expression of genes that are implicated in MS pathogenesis, as well as those encoding myelin proteins and transcription factors regulating new myelin synthesis. Together, these data indicate that neuroimmune modulation via VNS may be a promising approach to treat MS, that not only ameliorates symptoms but potentially also promotes myelin repair (remyelination).


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Estimulação do Nervo Vago , Nervo Vago , Animais , Encefalomielite Autoimune Experimental/terapia , Encefalomielite Autoimune Experimental/imunologia , Ratos , Esclerose Múltipla/terapia , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Estimulação do Nervo Vago/métodos , Inflamação/terapia , Inflamação/patologia , Modelos Animais de Doenças , Feminino , Bainha de Mielina/metabolismo , Barreira Hematoencefálica
3.
Nanotheranostics ; 8(4): 427-441, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38961889

RESUMO

Background: The blood-brain barrier (BBB) is a major bottleneck in delivering therapeutics to the brain. Treatment strategies to transiently open this barrier include focused ultrasound combined with intravenously injected microbubbles (FUS+MB) and targeting of molecules that regulate BBB permeability. Methods: Here, we investigated BBB opening mediated by the claudin-5 binder cCPEm (a microorganismal toxin in a truncated form) and FUS+MB at a centre frequency of 1 MHz, assessing dextran uptake, broadband emission, and endogenous immunoglobulin G (IgG) extravasation. Results: FUS+MB-induced BBB opening was detectable at a pressure ≥0.35 MPa when assessed for leakage of 10 and 70 kDa dextran, and at ≥0.2 MPa for uptake of endogenous IgG. Treating mice with 20 mg/kg cCPEm failed to open the BBB, and pre-treatment with cCPEm followed by FUS+MB at 0.2 and 0.3 MPa did not overtly increase BBB opening compared to FUS+MB alone. Using passive cavitation detection (PCD), we found that broadband emission correlated with the peak negative pressure (PNP) and dextran leakage, indicating the possibility of using broadband emission for developing a feedback controller to monitor BBB opening. Conclusions: Together, our study highlights the challenges in developing combinatorial approaches to open the BBB and presents an additional IgG-based histological detection method for BBB opening.


Assuntos
Barreira Hematoencefálica , Claudina-5 , Microbolhas , Animais , Barreira Hematoencefálica/metabolismo , Camundongos , Claudina-5/metabolismo , Imunoglobulina G/metabolismo , Ondas Ultrassônicas , Camundongos Endogâmicos C57BL , Dextranos/química , Dextranos/farmacocinética
4.
J Nanobiotechnology ; 22(1): 393, 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38965602

RESUMO

BACKGROUND: The therapeutic strategies for acute ischemic stroke were faced with substantial constraints, emphasizing the necessity to safeguard neuronal cells during cerebral ischemia to reduce neurological impairments and enhance recovery outcomes. Despite its potential as a neuroprotective agent in stroke treatment, Chikusetsu saponin IVa encounters numerous challenges in clinical application. RESULT: Brain-targeted liposomes modified with THRre peptides showed substantial uptake by bEnd. 3 and PC-12 cells and demonstrated the ability to cross an in vitro blood-brain barrier model, subsequently accumulating in PC-12 cells. In vivo, they could significantly accumulate in rat brain. Treatment with C-IVa-LPs-THRre notably reduced the expression of proteins in the P2RX7/NLRP3/Caspase-1 pathway and inflammatory factors. This was evidenced by decreased cerebral infarct size and improved neurological function in MCAO rats. CONCLUSION: The findings indicate that C-IVa-LPs-THRre could serve as a promising strategy for targeting cerebral ischemia. This approach enhances drug concentration in the brain, mitigates pyroptosis, and improves the neuroinflammatory response associated with stroke.


Assuntos
Barreira Hematoencefálica , AVC Isquêmico , Lipossomos , Fármacos Neuroprotetores , Piroptose , Ratos Sprague-Dawley , Saponinas , Animais , Saponinas/farmacologia , Saponinas/química , Piroptose/efeitos dos fármacos , Ratos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Lipossomos/química , Masculino , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/química , Células PC12 , Ácido Oleanólico/farmacologia , Ácido Oleanólico/química , Ácido Oleanólico/análogos & derivados , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Peptídeos/química , Peptídeos/farmacologia , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo
5.
Cell Mol Life Sci ; 81(1): 293, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38976012

RESUMO

The function of astrocytes in response to gut microbiota-derived signals has an important role in the pathophysiological processes of central nervous system (CNS) diseases. However, the specific effects of microbiota-derived metabolites on astrocyte activation have not been elucidated yet. Experimental autoimmune encephalomyelitis (EAE) was induced in female C57BL/6 mice as a classical MS model. The alterations of gut microbiota and the levels of short-chain fatty acids (SCFAs) were assessed after EAE induction. We observed that EAE mice exhibit low levels of Allobaculum, Clostridium_IV, Clostridium_XlVb, Lactobacillus genera, and microbial-derived SCFAs metabolites. SCFAs supplementation suppressed astrocyte activation by increasing the level of tryptophan (Trp)-derived AhR ligands that activating the AhR. The beneficial effects of SCFAs supplementation on the clinical scores, histopathological alterations, and the blood brain barrier (BBB)-glymphatic function were abolished by intracisterna magna injection of AAV-GFAP-shAhR. Moreover, SCFAs supplementation suppressed the loss of AQP4 polarity within astrocytes in an AhR-dependent manner. Together, SCFAs potentially suppresses astrocyte activation by amplifying Trp-AhR-AQP4 signaling in EAE mice. Our study demonstrates that SCFAs supplementation may serve as a viable therapy for inflammatory disorders of the CNS.


Assuntos
Aquaporina 4 , Astrócitos , Encefalomielite Autoimune Experimental , Ácidos Graxos Voláteis , Camundongos Endogâmicos C57BL , Receptores de Hidrocarboneto Arílico , Transdução de Sinais , Triptofano , Animais , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/metabolismo , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Ácidos Graxos Voláteis/farmacologia , Ácidos Graxos Voláteis/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Camundongos , Triptofano/metabolismo , Triptofano/farmacologia , Feminino , Transdução de Sinais/efeitos dos fármacos , Aquaporina 4/metabolismo , Aquaporina 4/genética , Microbioma Gastrointestinal/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos
6.
Sci Rep ; 14(1): 15338, 2024 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-38961135

RESUMO

Blood-brain barrier (BBB) disruption may contribute to cognitive decline, but questions remain whether this association is more pronounced for certain brain regions, such as the hippocampus, or represents a whole-brain mechanism. Further, whether human BBB leakage is triggered by excessive vascular pulsatility, as suggested by animal studies, remains unknown. In a prospective cohort (N = 50; 68-84 years), we used contrast-enhanced MRI to estimate the permeability-surface area product (PS) and fractional plasma volume ( v p ), and 4D flow MRI to assess cerebral arterial pulsatility. Cognition was assessed by the Montreal Cognitive Assessment (MoCA) score. We hypothesized that high PS would be associated with high arterial pulsatility, and that links to cognition would be specific to hippocampal PS. For 15 brain regions, PS ranged from 0.38 to 0.85 (·10-3 min-1) and v p from 0.79 to 1.78%. Cognition was related to PS (·10-3 min-1) in hippocampus (ß = - 2.9; p = 0.006), basal ganglia (ß = - 2.3; p = 0.04), white matter (ß = - 2.6; p = 0.04), whole-brain (ß = - 2.7; p = 0.04) and borderline-related for cortex (ß = - 2.7; p = 0.076). Pulsatility was unrelated to PS for all regions (p > 0.19). Our findings suggest PS-cognition links mainly reflect a whole-brain phenomenon with only slightly more pronounced links for the hippocampus, and provide no evidence of excessive pulsatility as a trigger of BBB disruption.


Assuntos
Barreira Hematoencefálica , Cognição , Imageamento por Ressonância Magnética , Humanos , Barreira Hematoencefálica/diagnóstico por imagem , Idoso , Masculino , Feminino , Cognição/fisiologia , Idoso de 80 Anos ou mais , Fluxo Pulsátil , Artérias Cerebrais/diagnóstico por imagem , Artérias Cerebrais/fisiologia , Estudos Prospectivos , Hipocampo/diagnóstico por imagem , Hipocampo/fisiologia , Encéfalo/diagnóstico por imagem , Encéfalo/fisiologia , Encéfalo/irrigação sanguínea , Disfunção Cognitiva/fisiopatologia , Disfunção Cognitiva/diagnóstico por imagem
7.
Biomolecules ; 14(6)2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38927100

RESUMO

Glaesserella parasuis (G. parasuis) causes serious inflammation and meningitis in piglets. Quercetin has anti-inflammatory and anti-bacterial activities; however, whether quercetin can alleviate brain inflammation and provide protective effects during G. parasuis infection has not been studied. Here, we established a mouse model of G. parasuis infection in vivo and in vitro to investigate transcriptome changes in the mouse cerebrum and determine the protective effects of quercetin on brain inflammation and blood-brain barrier (BBB) integrity during G. parasuis infection. The results showed that G. parasuis induced brain inflammation, destroyed BBB integrity, and suppressed PI3K/Akt/Erk signaling-pathway activation in mice. Quercetin decreased the expression of inflammatory cytokines (Il-18, Il-6, Il-8, and Tnf-α) and BBB-permeability marker genes (Mmp9, Vegf, Ang-2, and Et-1), increased the expression of angiogenetic genes (Sema4D and PlexinB1), reduced G. parasuis-induced tight junction disruption, and reactivated G. parasuis-induced suppression of the PI3K/Akt/Erk signaling pathway in vitro. Thus, we concluded that quercetin may protect BBB integrity via the PI3K/Akt/Erk signaling pathway during G. parasuis infection. This was the first attempt to explore the protective effects of quercetin on brain inflammation and BBB integrity in a G. parasuis-infected mouse model. Our findings indicated that quercetin is a promising natural agent for the prevention and treatment of G. parasuis infection.


Assuntos
Barreira Hematoencefálica , Modelos Animais de Doenças , Sistema de Sinalização das MAP Quinases , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Quercetina , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Quercetina/farmacologia , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Meningite/microbiologia , Meningite/tratamento farmacológico , Meningite/metabolismo , Infecções por Haemophilus/tratamento farmacológico , Infecções por Haemophilus/microbiologia , Transdução de Sinais/efeitos dos fármacos , Haemophilus parasuis/efeitos dos fármacos , Haemophilus parasuis/patogenicidade , Citocinas/metabolismo , Suínos
8.
Genes (Basel) ; 15(6)2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38927708

RESUMO

Extracellular vesicles (EVs) are "micro-shuttles" that play a role as mediators of intercellular communication. Cells release EVs into the extracellular environment in both physiological and pathological conditions and are involved in intercellular communication, due to their ability to transfer proteins, lipids, and nucleic acids, and in the modulation of the immune system and neuroinflammation. Because EVs can penetrate the blood-brain barrier and move from the central nervous system to the peripheral circulation, and vice versa, recent studies have shown a substantial role for EVs in several neurological diseases, including multiple sclerosis (MS). MS is a demyelinating disease where the main event is caused by T and B cells triggering an autoimmune reaction against myelin constituents. Recent research has elucidate the potential involvement of extracellular vesicles (EVs) in the pathophysiology of MS, although, to date, their potential role both as agents and therapeutic targets in MS is not fully defined. We present in this review a summary and comprehensive examination of EVs' involvement in the pathophysiology of multiple sclerosis, exploring their potential applications as biomarkers and indicators of therapy response.


Assuntos
Biomarcadores , Vesículas Extracelulares , Esclerose Múltipla , Humanos , Esclerose Múltipla/metabolismo , Esclerose Múltipla/terapia , Esclerose Múltipla/patologia , Vesículas Extracelulares/metabolismo , Animais , Barreira Hematoencefálica/metabolismo
9.
Int J Mol Sci ; 25(12)2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38928202

RESUMO

Blood-brain barrier (BBB) dysfunction is a key feature in neuroimmunological and neurodegenerative diseases. In this study, we developed a microfluidic human BBB-on-a-chip to model barrier dysfunction and immune cell migration using immortalized TY10 brain endothelial cells, pericytes, and astrocytes. It was found that immortalized TY10 brain endothelial cells developed a microvascular structure under flow. Pericytes were localized on the basal side surrounding the TY10 microvascular structure, showing an in vivo-like structure. Barrier integrity increased under co-culture with pericytes. In addition, both ethylenediaminetetraacetic acid (EDTA) and anti-Claudin-5 (CLDN5) neutralizing antibody caused a decrease in the transendothelial electrical resistance (TEER). EDTA caused the leakage of 20 kDa dextran, suggesting different effects on the BBB based on the mechanism of action, whereas anti-CLDN5 antibody did not cause leakage. In the tri-culture model, human T cells migrated through endothelial vessels towards basal C-X-C motif chemokine ligand 12 (CXCL12). The live-imaging analysis confirmed the extravasation of fluorescence-labelled T cells in a CXCL12-concentration- and time-dependent manner. Our BBB model had an in vivo-like structure and successfully represented barrier dysfunction and transendothelial T cell migration. In addition, our study suggests that the inhibition of CLDN5 attenuates the BBB in humans. This platform has various potential uses in relation to the BBB in both drug discovery research and in elucidating the mechanisms of central nervous system diseases.


Assuntos
Barreira Hematoencefálica , Movimento Celular , Células Endoteliais , Dispositivos Lab-On-A-Chip , Humanos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/efeitos dos fármacos , Descoberta de Drogas/métodos , Técnicas de Cocultura , Pericitos/metabolismo , Pericitos/efeitos dos fármacos , Claudina-5/metabolismo , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/efeitos dos fármacos
10.
Expert Opin Drug Deliv ; 21(5): 797-807, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38881261

RESUMO

BACKGROUND: Regadenoson, an agonist of adenosine A2 receptors, enables transient blood-brain barrier (BBB) disruption. The relevance of regadenoson as a pharmacological strategy for brain delivery was investigated using in vivo PET imaging in rats. RESEARCH DESIGN AND METHODS: Kinetic modeling of brain PET data was performed to estimate the impact of regadenoson (0.05 mg.kg-1, i.v.) on BBB permeation compared with control rats (n = 4-6 per group). Three radiolabeled compounds of different sizes, which do not cross the intact BBB, were tested. RESULTS: Regadenoson significantly increased the BBB penetration (+116 ± 13%, p < 0.001) of [18F]2-deoxy-2-fluoro-D-sorbitol ([18F]FDS, MW = 183 Da), a small-molecule marker of BBB permeability. The magnitude of the effect was different across brain regions, with a maximum increase in the striatum. Recovery of BBB integrity was observed 30 min after regadenoson injection. Regadenoson also increased the brain penetration (+72 ± 45%, p < 0.05) of a radiolabeled nanoparticle [89Zr]AGuIX (MW = 9 kDa). However, the brain kinetics of a monoclonal antibody ([89Zr]mAb, MW = 150 kDa) remained unchanged (p > 0.05). CONCLUSIONS: PET imaging showed the features and limitations of BBB disruption induced by regadenoson in terms of extent, regional distribution, and reversibility. Nevertheless, regadenoson enables the brain delivery of small molecules or nanoparticles in rats.


Assuntos
Agonistas do Receptor A2 de Adenosina , Barreira Hematoencefálica , Encéfalo , Tomografia por Emissão de Pósitrons , Purinas , Pirazóis , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Purinas/farmacologia , Purinas/administração & dosagem , Purinas/farmacocinética , Pirazóis/farmacologia , Pirazóis/administração & dosagem , Pirazóis/farmacocinética , Ratos , Tomografia por Emissão de Pósitrons/métodos , Encéfalo/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Masculino , Agonistas do Receptor A2 de Adenosina/farmacologia , Agonistas do Receptor A2 de Adenosina/administração & dosagem , Sistemas de Liberação de Medicamentos , Nanopartículas , Ratos Sprague-Dawley , Permeabilidade , Radioisótopos de Flúor , Ratos Wistar
11.
Brain Res Bull ; 214: 110987, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38830487

RESUMO

In modern war or daily life, blast-induced traumatic brain injury (bTBI) is a growing health concern. Our previous studies demonstrated that inflammation was one of the main features of bTBI, and CD28-activated T cells play a central role in inflammation. However, the mechanism of CD28 in bTBI remains to be elucidated. In this study, traumatic brain injury model induced by chest blast exposure in male mice was established, and the mechanism of CD28 in bTBI was studied by elisa, immunofluorescence staining, flow cytometry analysis and western blot. After exposure to chest shock wave, the inflammatory factors IL-4, IL-6 and HMGB1 in serum were increased, and CD3+ T cells, CD4+ and CD8+ T cell subsets in the lung were activated. In addition, chest blast exposure resulted in impaired spatial learning and memory ability, disruption of the blood-brain barrier (BBB), and the expression of Tau, p-tau, S100ß and choline acetyltransferase were increased. The results indicated that genetic knockdown of CD28 could inhibit inflammatory cell infiltration, as well as the activation of CD3+ T cells, CD4+ and CD8+ T cell subsets in the lung, improve spatial learning and memory ability, and ameliorate BBB disruption and hippocampal neuron damage. Moreover, genetic knockdown of CD28 could reduce the expression of p-PI3K, p-AKT and NF-κB. In conclusion, chest blast exposure could lead to bTBI, and attenuate bTBI via the PI3K/AKT/NF-κB signaling pathway in male mice. This study provides new targets for the prevention and treatment of veterans with bTBI.


Assuntos
Traumatismos por Explosões , Lesões Encefálicas Traumáticas , Antígenos CD28 , Camundongos Endogâmicos C57BL , NF-kappa B , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Animais , Masculino , Lesões Encefálicas Traumáticas/metabolismo , Antígenos CD28/metabolismo , Transdução de Sinais/fisiologia , Traumatismos por Explosões/complicações , Traumatismos por Explosões/metabolismo , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Modelos Animais de Doenças , Barreira Hematoencefálica/metabolismo , Traumatismos Torácicos/complicações
12.
Cells ; 13(12)2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38920629

RESUMO

The world of cancer treatment is evolving rapidly and has improved the prospects of many cancer patients. Yet, there are still many cancers where treatment prospects have not (or hardly) improved. Glioblastoma is the most common malignant primary brain tumor, and even though it is sensitive to many chemotherapeutics when tested under laboratory conditions, its clinical prospects are still very poor. The blood-brain barrier (BBB) is considered at least partly responsible for the high failure rate of many promising treatment strategies. We describe the workings of the BBB during healthy conditions and within the glioblastoma environment. How the BBB acts as a barrier for therapeutic options is described as well as various approaches developed and tested for passing or opening the BBB, with the ultimate aim to allow access to brain tumors and improve patient perspectives.


Assuntos
Barreira Hematoencefálica , Neoplasias Encefálicas , Sistemas de Liberação de Medicamentos , Glioblastoma , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/métodos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Antineoplásicos/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/administração & dosagem , Animais
13.
Sci Rep ; 14(1): 14438, 2024 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-38910141

RESUMO

Blood-brain barrier disruption is a critical pathological event in the progression of ischemic stroke (IS). Most studies regarding the therapeutic potential of neferine (Nef) on IS have focused on neuroprotective effect. However, whether Nef attenuates BBB disruption during IS is unclear. We here used mice underwent transient middle cerebral artery occlusion (tMCAO) in vivo and bEnd.3 cells exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) injury in vitro to simulate cerebral ischemia. We showed that Nef reduced neurobehavioral dysfunction and protected brain microvascular endothelial cells and BBB integrity. Molecular docking, short interfering (Si) RNA and plasmid transfection results showed us that PGC-1α was the most binding affinity of biological activity protein for Nef. And verification experiments were showed that Nef upregulated PGC-1α expression to reduce mitochondrial oxidative stress and promote TJ proteins expression, further improves the integrity of BBB in mice. Intriguingly, our study showed that neferine is a natural PGC-1α activator and illustrated the mechanism of specific binding site. Furthermore, we have demonstrated Nef reduced mitochondria oxidative damage and ameliorates endothelial inflammation by inhibiting pyroptosis to improve BBB permeability through triggering a cascade reaction of PGC-1α via regulation of PGC-1α/NLRP3/GSDMD signaling pathway to maintain the integrity of BBB in ischemia/reperfusion injury.


Assuntos
Benzilisoquinolinas , Barreira Hematoencefálica , Células Endoteliais , AVC Isquêmico , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Piroptose , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Piroptose/efeitos dos fármacos , Camundongos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , AVC Isquêmico/metabolismo , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/patologia , Células Endoteliais/metabolismo , Células Endoteliais/efeitos dos fármacos , Benzilisoquinolinas/farmacologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Fármacos Neuroprotetores/farmacologia
14.
Sci Rep ; 14(1): 14535, 2024 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-38914585

RESUMO

The rapid perfusion of cerebral arteries leads to a significant increase in intracranial blood volume, exposing patients with traumatic brain injury to the risk of diffuse brain swelling or malignant brain herniation during decompressive craniectomy. The microcirculation and venous system are also involved in this process, but the precise mechanisms remain unclear. A physiological model of extremely high intracranial pressure was created in rats. This development triggered the TNF-α/NF-κB/iNOS axis in microglia, and released many inflammatory factors and reactive oxygen species/reactive nitrogen species, generating an excessive amount of peroxynitrite. Subsequently, the capillary wall cells especially pericytes exhibited severe degeneration and injury, the blood-brain barrier was disrupted, and a large number of blood cells were deposited within the microcirculation, resulting in a significant delay in the recovery of the microcirculation and venous blood flow compared to arterial flow, and this still persisted after decompressive craniectomy. Infliximab is a monoclonal antibody bound to TNF-α that effectively reduces the activity of TNF-α/NF-κB/iNOS axis. Treatment with Infliximab resulted in downregulation of inflammatory and oxidative-nitrative stress related factors, attenuation of capillary wall cells injury, and relative reduction of capillary hemostasis. These improved the delay in recovery of microcirculation and venous blood flow.


Assuntos
Hipertensão Intracraniana , Estresse Oxidativo , Animais , Ratos , Hipertensão Intracraniana/etiologia , Hipertensão Intracraniana/tratamento farmacológico , Masculino , Fator de Necrose Tumoral alfa/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Microcirculação , Circulação Cerebrovascular , Ratos Sprague-Dawley , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Infliximab/farmacologia , Infliximab/uso terapêutico , Modelos Animais de Doenças , Barreira Hematoencefálica/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Microglia/metabolismo
15.
Biopharm Drug Dispos ; 45(3): 138-148, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38823029

RESUMO

Bumetanide is used widely as a tool and off-label treatment to inhibit the Na-K-2Cl cotransporter NKCC1 in the brain and thereby to normalize intra-neuronal chloride levels in several brain disorders. However, following systemic administration, bumetanide only poorly penetrates into the brain parenchyma and does not reach levels sufficient to inhibit NKCC1. The low brain penetration is a consequence of both the high ionization rate and plasma protein binding, which restrict brain entry by passive diffusion, and of brain efflux transport. In previous studies, bumetanide was determined in the whole brain or a few brain regions, such as the hippocampus. However, the blood-brain barrier and its efflux transporters are heterogeneous across brain regions, so it cannot be excluded that bumetanide reaches sufficiently high brain levels for NKCC1 inhibition in some discrete brain areas. Here, bumetanide was determined in 14 brain regions following i.v. administration of 10 mg/kg in rats. Because bumetanide is much more rapidly eliminated by rats than humans, its metabolism was reduced by pretreatment with piperonyl butoxide. Significant, up to 5-fold differences in regional bumetanide levels were determined with the highest levels in the midbrain and olfactory bulb and the lowest levels in the striatum and amygdala. Brain:plasma ratios ranged between 0.004 (amygdala) and 0.022 (olfactory bulb). Regional brain levels were significantly correlated with local cerebral blood flow. However, regional bumetanide levels were far below the IC50 (2.4 µM) determined previously for rat NKCC1. Thus, these data further substantiate that the reported effects of bumetanide in rodent models of brain disorders are not related to NKCC1 inhibition in the brain.


Assuntos
Encéfalo , Bumetanida , Animais , Bumetanida/farmacologia , Bumetanida/farmacocinética , Bumetanida/administração & dosagem , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Masculino , Ratos , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacocinética , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Inibidores de Simportadores de Cloreto de Sódio e Potássio/administração & dosagem , Ratos Sprague-Dawley , Distribuição Tecidual , Membro 2 da Família 12 de Carreador de Soluto/metabolismo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos
16.
Chem Commun (Camb) ; 60(53): 6793-6796, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38869018

RESUMO

We report an integrated ratiometric lysosomal nitric oxide (NO) nanoprobe based on engineered semiconducting polymer dots (Pdots), LyNO-Pdots, which consist of a newly designed NO-responsive dye, a fluorescent conjugated polymer and two functional polymers. The developed probe LyNO-Pdots exhibit high specificity and stability, good photostability and favorable blood-brain barrier (BBB) penetration ability. The LyNO-Pdots are successfully applied to ratiometric imaging of lysosomal NO variations in brain-derived endothelial cells, brain tissues and mice brains with Alzheimer's disease (AD). The results demonstrate that the NO content in the brains of AD mice is considerably higher than that in normal mice.


Assuntos
Doença de Alzheimer , Encéfalo , Corantes Fluorescentes , Lisossomos , Óxido Nítrico , Imagem Óptica , Animais , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/metabolismo , Lisossomos/química , Lisossomos/metabolismo , Camundongos , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico/análise , Corantes Fluorescentes/química , Corantes Fluorescentes/síntese química , Humanos , Polímeros/química , Barreira Hematoencefálica/metabolismo , Pontos Quânticos/química
17.
Nat Commun ; 15(1): 5000, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38866763

RESUMO

To date, earlier diagnosis of Alzheimer's disease (AD) is still challenging. Recent studies revealed the elevated expression of connective tissue growth factor (CTGF) in AD brain is an upstream regulator of amyloid-beta (Aß) plaque, thus CTGF could be an earlier diagnostic biomarker of AD than Aß plaque. Herein, we develop a peptide-coated gold nanocluster that specifically targets CTGF with high affinity (KD ~ 21.9 nM). The probe can well penetrate the blood-brain-barrier (BBB) of APP/PS1 transgenic mice at early-stage (earlier than 3-month-old) in vivo, allowing non-invasive NIR-II imaging of CTGF when there is no appearance of Aß plaque deposition. Notably, this probe can also be applied to measuring CTGF on postmortem brain sections by multimodal analysis, including fluorescence imaging, peroxidase-like chromogenic imaging, and ICP-MS quantitation, which enables distinguishment between the brains of AD patients and healthy people. This probe possesses great potential for precise diagnosis of earlier AD before Aß plaque formation.


Assuntos
Doença de Alzheimer , Encéfalo , Fator de Crescimento do Tecido Conjuntivo , Camundongos Transgênicos , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Animais , Humanos , Camundongos , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Encéfalo/patologia , Ouro/química , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/diagnóstico por imagem , Placa Amiloide/diagnóstico por imagem , Placa Amiloide/metabolismo , Nanopartículas Metálicas/química , Modelos Animais de Doenças , Peptídeos beta-Amiloides/metabolismo , Feminino , Masculino , Imagem Multimodal/métodos , Biomarcadores/metabolismo , Imagem Óptica/métodos
18.
CNS Neurosci Ther ; 30(6): e14796, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38867395

RESUMO

AIMS: The extent of perihematomal edema following intracerebral hemorrhage (ICH) significantly impacts patient prognosis, and disruption of the blood-brain barrier (BBB) exacerbates perihematomal edema. However, the role of peripheral IL-10 in mitigating BBB disruption through pathways that link peripheral and central nervous system signals remains poorly understood. METHODS: Recombinant IL-10 was administered to ICH model mice via caudal vein injection, an IL-10-inhibiting adeno-associated virus and an IL-10 receptor knockout plasmid were delivered intraventricularly, and neurobehavioral deficits, perihematomal edema, BBB disruption, and the expression of JAK1 and STAT3 were evaluated. RESULTS: Our study demonstrated that the peripheral cytokine IL-10 mitigated BBB breakdown, perihematomal edema, and neurobehavioral deficits after ICH and that IL-10 deficiency reversed these effects, likely through the IL-10R/JAK1/STAT3 signaling pathway. CONCLUSIONS: Peripheral IL-10 has the potential to reduce BBB damage and perihematomal edema following ICH and improve patient prognosis.


Assuntos
Edema Encefálico , Hemorragia Cerebral , Interleucina-10 , Janus Quinase 1 , Receptores de Interleucina-10 , Fator de Transcrição STAT3 , Transdução de Sinais , Animais , Fator de Transcrição STAT3/metabolismo , Hemorragia Cerebral/complicações , Hemorragia Cerebral/tratamento farmacológico , Hemorragia Cerebral/metabolismo , Edema Encefálico/etiologia , Edema Encefálico/tratamento farmacológico , Janus Quinase 1/metabolismo , Janus Quinase 1/antagonistas & inibidores , Interleucina-10/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo
19.
J Neuroinflammation ; 21(1): 155, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38872149

RESUMO

Activation of the kallikrein-kinin system promotes vascular leakage, inflammation, and neurodegeneration in ischemic stroke. Inhibition of plasma kallikrein (PK) - a key component of the KKS - in the acute phase of ischemic stroke has been reported to reduce thrombosis, inflammation, and damage to the blood-brain barrier. However, the role of PK during the recovery phase after cerebral ischemia is unknown. To this end, we evaluated the effect of subacute PK inhibition starting from day 3 on the recovery process after transient middle artery occlusion (tMCAO). Our study demonstrated a protective effect of PK inhibition by reducing infarct volume and improving functional outcome at day 7 after tMCAO. In addition, we observed reduced thrombus formation in cerebral microvessels, fewer infiltrated immune cells, and an improvement in blood-brain barrier integrity. This protective effect was facilitated by promoting tight junction reintegration, reducing detrimental matrix metalloproteinases, and upregulating regenerative angiogenic markers. Our findings suggest that PK inhibition in the subacute phase might be a promising approach to accelerate the post-stroke recovery process.


Assuntos
Calicreína Plasmática , Recuperação de Função Fisiológica , Animais , Recuperação de Função Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/fisiologia , Masculino , Calicreína Plasmática/antagonistas & inibidores , Calicreína Plasmática/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Infarto da Artéria Cerebral Média , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Trombose , AVC Isquêmico/tratamento farmacológico , Inflamação
20.
Int J Nanomedicine ; 19: 6177-6199, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38911498

RESUMO

Purpose: Ginsenoside Rg3 (Rg3) and Panax notoginseng saponins (PNS) can be used for ischemic stroke treatment, however, the lack of targeting to the ischemic region limits the therapeutic effect. To address this, we leveraged the affinity of macrophage membrane proteins for inflamed brain microvascular endothelial cells to develop a macrophage membrane-cloaked liposome loaded with Rg3 and PNS (MM-Lip-Rg3/PNS), which can precisely target brain lesion region through intranasal administration. Methods: MM-Lip-Rg3/PNS was prepared by co-extrusion method and was performed by characterization, stability, surface protein, and morphology. The cellular uptake, immune escape ability, and blood-brain barrier crossing ability of MM-Lip-Rg3/PNS were studied in vitro. The in vivo brain targeting, biodistribution and anti-ischemic efficacy of MM-Lip-Rg3/PNS were evaluated in MACO rats, and we determined the diversity of the nasal brain pathway through the olfactory nerve blockade model in rats. Finally, the pharmacokinetics and brain targeting index of MM-Lip-Rg3/PNS were investigated. Results: Our results indicated that MM-Lip-Rg3/PNS was spherical with a shell-core structure. MM-Lip-Rg3/PNS can avoid mononuclear phagocytosis, actively bind to inflammatory endothelial cells, and have the ability to cross the blood-brain barrier. Moreover, MM-Lip-Rg3/PNS could specifically target ischemic sites, even microglia, increase the cumulative number of drugs in the brain, improve the inflammatory environment of the brain, and reduce the infarct size. By comparing olfactory nerve-blocking rats with normal rats, it was found that there are direct and indirect pathways for nasal entry into the brain. Pharmacokinetics demonstrated that MM-Lip-Rg3/PNS exhibited stronger brain targeting and prolonged drug half-life. Conclusion: MM-Lip-Rg3/PNS might contribute to the accumulation of Rg3 and PNS in the ischemic brain area to improve treatment efficacy. This biomimetic nano-drug delivery system provides a new and promising strategy for the treatment of ischemic stroke.


Assuntos
Administração Intranasal , Barreira Hematoencefálica , Ginsenosídeos , AVC Isquêmico , Lipossomos , Macrófagos , Animais , Lipossomos/química , AVC Isquêmico/tratamento farmacológico , Ratos , Masculino , Ginsenosídeos/farmacocinética , Ginsenosídeos/química , Ginsenosídeos/administração & dosagem , Ginsenosídeos/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/métodos , Ratos Sprague-Dawley , Distribuição Tecidual , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Materiais Biomiméticos/administração & dosagem , Saponinas/farmacocinética , Saponinas/química , Saponinas/administração & dosagem , Saponinas/farmacologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...