Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Mol Pharm ; 21(7): 3684-3692, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38899595

RESUMO

Early detection of pulmonary fibrosis is a critical yet insufficiently met clinical necessity. This study evaluated the effectiveness of FAPI-LM3, a 68Ga-radiolabeled heterobivalent molecular probe that targets fibroblast activating protein (FAP) and somatostatin receptor 2 (SSTR2), in the early detection of pulmonary fibrosis, leveraging its potential for early disease identification. A bleomycin-induced early pulmonary fibrosis model was established in C57BL/6 mice for 7 days. FAP and SSTR2 expression levels were quantitatively assessed in human idiopathic pulmonary fibrosis lung tissue samples and bleomycin-treated mouse lung tissues by using western blotting, real-time quantitative PCR (RT-qPCR), and immunofluorescence techniques. The diagnostic performance of FAPI-LM3 was investigated by synthesizing monomeric radiotracers 68Ga-FAPI-46 and 68Ga-DOTA-LM3 alongside the heterobivalent probe 68Ga-FAPI-LM3. These imaging radiopharmaceuticals were used in small-animal PET to compare their uptake in fibrotic and normal lung tissues. Results indicated significant upregulation of FAP and SSTR2 at both RNA and protein levels in fibrotic lung tissues compared with that in normal controls. PET imaging demonstrated significantly enhanced uptake of the 68Ga-FAPI-LM3 probe in fibrotic lung tissues, with superior visual effects compared to monomeric tracers. At 60 min postinjection, early stage fibrotic tissues (day 7) demonstrated low-to-medium uptake of monomeric probes, including 68Ga-DOTA-LM3 (0.45 ± 0.04% ID/g) and 68Ga-FAPI-46 (0.78 ± 0.09% ID/g), whereas the uptake of the heterobivalent probe 68Ga-FAPI-LM3 (1.90 ± 0.10% ID/g) was significantly higher in fibrotic lesions than in normal lung tissue. Blockade experiments confirmed the specificity of 68Ga-FAPI-LM3 uptake, which was attributed to synergistic targeting of FAP and SSTR2. This study demonstrates the potential of 68Ga-FAPI-LM3 for early pulmonary fibrosis detection via molecular imaging, offering significant benefits over monomeric tracers 68Ga-FAPI-46 and 68Ga-DOTA-LM3. This strategy offers new possibilities for noninvasive and precise early detection of pulmonary fibrosis.


Assuntos
Radioisótopos de Gálio , Camundongos Endogâmicos C57BL , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Receptores de Somatostatina , Animais , Camundongos , Receptores de Somatostatina/metabolismo , Humanos , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/farmacocinética , Compostos Radiofarmacêuticos/química , Fibrose Pulmonar/diagnóstico por imagem , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/induzido quimicamente , Pulmão/diagnóstico por imagem , Pulmão/patologia , Pulmão/metabolismo , Masculino , Bleomicina , Endopeptidases/metabolismo , Modelos Animais de Doenças , Feminino , Fibrose Pulmonar Idiopática/diagnóstico por imagem , Fibrose Pulmonar Idiopática/metabolismo , Proteínas de Membrana/metabolismo , Serina Endopeptidases/metabolismo , Quinolinas
2.
Pharmacol Res ; 141: 530-540, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30660821

RESUMO

Glial activation and scar formation impede the neurological function recovery after cerebral ischemia. Oleoylethanolamide (OEA), a bioactive lipid mediator, shows neuroprotection against acute brain ischemia, however, its long-term effect, especially on glial scar formation, has not been characterized. In this research, we investigate the effect of OEA on glial activation and scar formation after cerebral ischemia in vitro and in vivo experiments. Glial scar formation in vitro model was induced by transforming growth factor ß1 (TGF-ß1) in C6 glial cell culture, and experiment model in vivo was induced by middle cerebral artery occlusion (MCAO) in mice. The protein expressions of the markers of glial activation (S100ß, GFAP, or pSmads) and glial scar (neurocan) were detected by Western blot and/or immunofluorescence staining; To evaluate the role of PPARɑ in the effect of OEA on glial activation, the PPARɑ antagonist GW6471 was used. Behavior tests were used to assay the effect of OEA on motor function recovery 14 days after brain ischemia in mice. Our results show that OEA (10-50 µM) concentration-dependently inhibited the upregulation of S100ß, GFAP, pSmads and neurocan induced by TGF-ß1 in C6 glial cells. At the same time, OEA promoted the protein expression and nuclear transportation of PPARɑ in glial cells. PPARα antagonist GW6471 abolished the effect of OEA on glial activation. In addition, we found that delay administration of OEA inhibited the astrocyte activation and promoted the recovery of motor function after brain ischemia in mice. These results indicate that OEA may be developed into a new candidate for attenuating astrocytic scar formation and improving motor function after ischemic stroke.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Endocanabinoides/uso terapêutico , Neuroglia/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Ácidos Oleicos/uso terapêutico , PPAR alfa/metabolismo , Animais , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Linhagem Celular , Endocanabinoides/farmacologia , Força da Mão , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Camundongos , Neuroglia/metabolismo , Neuroglia/patologia , Fármacos Neuroprotetores/farmacologia , Ácidos Oleicos/farmacologia , Ratos , Recuperação de Função Fisiológica , Caminhada
3.
CNS Neurosci Ther ; 30(2): e14363, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37469216

RESUMO

AIMS: Acute kidney injury (AKI) has been associated with a variety of neurological problems, while the neurobiological mechanism remains unclear. In the present study, we utilized resting-state functional magnetic resonance imaging (rs-fMRI) to detect brain injury at an early stage and investigated the impact of microglia on the neuropathological mechanism of AKI. METHODS: Rs-fMRI data were collected from AKI rats and the control group with a 9.4-Tesla scanner at 24, 48, and 72 h post administration of contrast medium or saline. The amplitude of low-frequency fluctuations (ALFF) was then compared across the groups at each time course. Additionally, flow cytometry and SMART-seq2 were employed to evaluate microglia. Furthermore, pathological staining and Western blot were used to analyze the samples. RESULTS: MRI results revealed that AKI led to a decreased ALFF in the hippocampus, particularly in the 48 h and 72 h groups. Additionally, western blot suggested that AKI-induced the neuronal apoptosis at 48 h and 72 h. Flow cytometry and confocal microscopy images demonstrated that AKI activated the aggregation of microglia into neurons at 24 h, with a strong upregulation of M1 polarization at 48 h and peaking at 72 h, accompanying with the release of proinflammatory cytokines. The ALFF value was strongly correlated with the proportion of microglia (|r| > 0.80, p < 0.001). CONCLUSIONS: Our study demonstrated that microglia aggregation and inflammatory factor upregulation are significant mechanisms of AKI-induced neuronal apoptosis. We used fMRI to detect the alterations in hippocampal function, which may provide a noninvasive method for the early detection of brain injury after AKI.


Assuntos
Injúria Renal Aguda , Lesões Encefálicas , Ratos , Animais , Microglia , Hipocampo/diagnóstico por imagem , Apoptose , Injúria Renal Aguda/diagnóstico por imagem
4.
Mater Today Bio ; 26: 101094, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38854952

RESUMO

Cerebral ischemia-reperfusion injury (CIRI) is a major challenge to neuronal survival in acute ischemic stroke (AIS). However, effective neuroprotective agents remain to be developed for the treatment of CIRI. In this work, we have developed an Anti-TRAIL protein-modified and indocyanine green (ICG)-responsive nanoagent (Anti-TRAIL-ICG) to target ischemic areas and then reduce CIRI and rescue the ischemic penumbra. In vitro and in vivo experiments have demonstrated that the carrier-free nanoagent can enhance drug transport across the blood-brain barrier (BBB) in stroke mice, exhibiting high targeting ability and good biocompatibility. Anti-TRAIL-ICG nanoagent played a better neuroprotective role by reducing apoptosis and ferroptosis, and significantly improved ischemia-reperfusion injury. Moreover, the multimodal imaging platform enables the dynamic in vivo examination of multiple morphofunctional information, so that the dynamic molecular events of nanoagent can be detected continuously and in real time for early treatment in transient middle cerebral artery occlusion (tMCAO) models. Furthermore, it has been found that Anti-TRAIL-ICG has great potential in the functional reconstruction of neurovascular networks through optical coherence tomography angiography (OCTA). Taken together, our work effectively alleviates CIRI after stoke by blocking multiple cell death pathways, which offers an innovative strategy for harnessing the apoptosis and ferroptosis against CIRI.

5.
Biomedicines ; 11(3)2023 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-36979952

RESUMO

Astrocyte inflammation activation is an important cause that hinders the recovery of motor function after cerebral ischemia. However, its molecular mechanism has not yet been clearly clarified. The peroxisome proliferator-activated receptor α (PPARα) is a ligand-activated nuclear transcriptional factor. This study aims to further clarify the role of PPARα in astrocyte inflammation activation after cerebral ischemia and to explore the underlying mechanism. Astrocyte activation was induced in an in vivo model by transient middle cerebral artery occlusion (tMCAO) in mice. The in vitro model was induced by an oxygen-glucose deprivation/reoxygenation (OGD/R) in a primary culture of mouse astrocyte. PPARα-deficient mice were used to observe the effects of PPARα on astrocyte activation and autophagic flux. Our results showed that PPARα was mainly expressed in activated astrocytes during the chronic phase of brain ischemia and PPARα dysfunction promoted astrocyte inflammatory activation. After cerebral ischemia, the expressions of LC3-II/I and p62 both increased. Autophagic vesicle accumulation was observed by electron microscopy in astrocytes, and the block of autophagic flux was indicated by an mRFP-GFP-LC3 adenovirus infection assay. A PPARα deficit aggravated the autophagic flux block, while PPARα activation preserved the lysosome function and restored autophagic flux in astrocytes after OGD/R. The autophagic flux blocker bafilomycin A1 and chloroquine antagonized the effect of the PPARα agonist on astrocyte activation inhibition. This study identifies a potentially novel function of PPARα in astrocyte autophagic flux and suggests a therapeutic target for the prevention and treatment of chronic brain ischemic injury.

6.
Eur J Pharmacol ; 957: 175982, 2023 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-37572942

RESUMO

Ischemic stroke is a leading cause of death and disability, and medical treatments for ischemic stroke are very limited. URB597 is a potent and selective inhibitor of fatty acid amide hydrolase (FAAH). However, the effect of URB597 on ischemic stroke and the underlying molecular mechanisms remain little known. In this study, focal cerebral ischemia was induced by transient middle cerebral artery occlusion in mice. Our results showed that URB597 dose-dependently improved neurological function and reduced brain infarct volume and brain edema 24 h after brain ischemia. The most effective dose was 1 mg/kg and the therapeutic time window was within 3 h after ischemic stroke. To further investigate the underlying mechanism, necroptosis and autophagy flux were detected by Western blot and/or immunofluorescence staining with or without chloroquine, an autophagic flux inhibitor. Our results showed that URB597 promoted autophagic flux and reduced neuronal necroptosis after brain ischemia and these effects could be abolished by chloroquine. In addition, we found that peroxisome proliferator-activated receptor α (PPARα) antagonist GW6471 partly abolished the effect of URB597 against brain ischemia and URB597 upregulated the expressions of PPARα. In conclusion, URB597 exerts a neuroprotective effect in a dose- and time-dependent manner, and this effect may be related to its restoration of autophagic flux and inhibition of neuronal necroptosis. PPARα is involved in the neuroprotective effect of URB597. This study provides novel evidence that URB597 may be a promising agent for the clinical treatment of ischemic stroke.


Assuntos
Lesões Encefálicas , Isquemia Encefálica , AVC Isquêmico , Fármacos Neuroprotetores , Ratos , Camundongos , Animais , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , PPAR alfa/metabolismo , Necroptose , Ratos Sprague-Dawley , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/tratamento farmacológico , Autofagia , Cloroquina/farmacologia , Cloroquina/uso terapêutico
7.
Biomed Pharmacother ; 107: 44-53, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30077837

RESUMO

Oxidative stress is an important cause of skin injury induced by UVB radiation. Moringa oleifera also known as horseradish tree or drumstick tree, have multiple nutraceutical or pharmacological functions. However, whether Moringa oleifera protects skin against oxidative stress injury remains unknown. To investigate the effects of the ethanol extract of Moringa oleifera stem (MSE) on skin oxidative stress injury and its molecular mechanism, we first determined the effect of MSE on epidermal oxidative stress injury induced by H2O2 in keratinocytes (HaCaT cells) and by UVB-radiation in mice. Then we investigated the effect of MSE on the enhancement of antioxidant system and activation of PPARα in vitro and in vivo. Furthermore, the flavonoids compositions in MSE were assayed by high-performance liquid chromatography (HPLC), and then molecular docking study was used to assess the major component in MSE to activate PPARα. Our results indicate that MSE (100-400 µg/mL) protected the epidemic cell against oxidative stress injury in vitro and topical treatment with MSE cream (6%) inhibit UVB-induced oxidative stress injury in the epidermis of the mouse skin. PPARα activation is involved in the protective effect of MSE. HPLC assay and molecular docking study indicated that rutin might be the main component in MSE to activate PPARα. These results confirm that MSE exerts the protective effect on oxidative stress induced skin keratinocytes injury. Moreover, the protective effect of MSE is mediated by enhancement of antioxidant defense systems and activation of PPARα in skin keratinocytes.


Assuntos
Antioxidantes/farmacologia , Queratinócitos/patologia , Moringa oleifera/química , Estresse Oxidativo/efeitos dos fármacos , PPAR alfa/metabolismo , Extratos Vegetais/farmacologia , Caules de Planta/química , Substâncias Protetoras/farmacologia , Animais , Catalase/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular , Citoproteção/efeitos dos fármacos , Epiderme/patologia , Epiderme/efeitos da radiação , Feminino , Humanos , Peróxido de Hidrogênio/toxicidade , Queratinócitos/efeitos dos fármacos , Queratinócitos/enzimologia , Camundongos Endogâmicos BALB C , Simulação de Acoplamento Molecular , Espécies Reativas de Oxigênio/metabolismo , Rutina/química , Rutina/farmacologia , Pele/patologia , Superóxido Dismutase/metabolismo , Raios Ultravioleta
8.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 23(1): 106-10, 2007 Feb.
Artigo em Zh | MEDLINE | ID: mdl-21171382

RESUMO

AIM: To explore the effect of sodium 4',7-bihydroxylisoflavone-sulfonate (SBIS) on gastric motility in rats and to analyse its mechanisms. METHODS: Using intraperitoneal (ip) injection and intracerebroventriular (icv) microinjection of drugs and recording the frequency and amplitude of contraction of gastric motility. RESULTS: (1) The injection (ip) of different doses of SBIS could decrease the amplitude of gastric motility, but it wasn't a dose-dependent effect. SBIS also had no effect on the frequency of contraction. (2) The ip injection of naloxone reversed the inhibitory effect of SBIS on the amplitude of gastric contraction. (3) The effect of SBIS could be increased by the ip injection of propranolol and be reversed by the ip injection of phentolamine. (4) After the ip injection of atropine, the effect of SBIS on gastric motility had not been changed remarkably. (5) Different doses of SBIS had been microinjected (icy), but only the small dose decreased the amplitude of gastric motility and also the frequency of contraction had not been markedly changed. CONCLUSION: Both the i.p. and icv injection of SBIS can inhibit the gastric motility. Its effect can be achieved at least not only by endogenous opioid peptide and its receptors, but also adrenergic neuron and its alpha-receptors. Adrenergic neuron and its beta-receptors are also involved in the modulating effect of SBIS.


Assuntos
Motilidade Gastrointestinal/efeitos dos fármacos , Isoflavonas/farmacologia , Animais , Masculino , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA