Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
1.
Drug Des Devel Ther ; 18: 767-780, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38495631

RESUMO

Purpose: Pulmonary arterial hypertension (PAH) is a devastating disease with little effective treatment. The proliferation of pulmonary artery smooth muscle cells (PASMCs) induced by the nuclear factor-κB (NF-κB) signaling activation plays a pivotal role in the pathogenesis of PAH. Forsythoside B (FTS•B) possesses inhibitory effect on NF-κB signaling pathway. The present study aims to explore the effects and mechanisms of FTS•B in PAH. Methods: Sprague-Dawley rats received monocrotaline (MCT) intraperitoneal injection to establish PAH model, and FTS•B was co-treated after MCT injection. Right ventricular hypertrophy and pulmonary artery pressure were measured by echocardiography and right heart catheterization, respectively. Histological alterations were detected by H&E staining and immunohistochemistry. FTS•B's role in PASMC proliferation and migration were evaluated by CCK-8 and wound healing assay. To investigate the underlying mechanisms, Western blotting, immunofluorescence staining and ELISA were conducted. The NF-κB activator PMA was used to investigate the role of NF-κB in FTS•B's protective effects against PAH. Results: FTS•B markedly alleviated MCT-induced vascular remodeling and pulmonary artery pressure, and improved right ventricular hypertrophy and survival. FTS•B also reversed PDGF-BB-induced PASMC proliferation and migration, decreased PCNA and CyclinD1 expression in vitro. The elevated levels of IL-1ß and IL-6 caused by MCT were decreased by FTS•B. Mechanistically, MCT-triggered phosphorylation of p65, IκBα, IKKα and IKKß was blunted by FTS•B. FTS•B also reversed MCT-induced nuclear translocation of p65. However, all these protective effects were blocked by PMA-mediated NF-κB activation. Conclusion: FTS•B effectively attenuates PAH by suppressing the NF-κB signaling pathway to attenuate vascular remodeling. FTS•B might be a promising drug candidate with clinical translational potential for the treatment of PAH.


Assuntos
Ácidos Cafeicos , Glucosídeos , Hipertensão Pulmonar , Hipertensão Arterial Pulmonar , Ratos , Animais , NF-kappa B/metabolismo , Monocrotalina/efeitos adversos , Ratos Sprague-Dawley , Remodelação Vascular , Hipertrofia Ventricular Direita/metabolismo , Hipertrofia Ventricular Direita/patologia , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/tratamento farmacológico , Transdução de Sinais
2.
Front Cardiovasc Med ; 11: 1364604, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38529329

RESUMO

Belonging to a lipid phosphatase family containing 16 members, myotubularin-related proteins (MTMRs) are widely expressed in a variety of tissues and organs. MTMRs preferentially hydrolyzes phosphatidylinositol 3-monophosphate and phosphatidylinositol (3,5) bis-phosphate to generate phosphatidylinositol and phosphatidylinositol 5-monophosphate, respectively. These phosphoinositides (PIPs) promote membrane degradation during autophagosome-lysosomal fusion and are also involved in various regulatory signal transduction. Based on the ability of modulating the levels of these PIPs, MTMRs exert physiological functions such as vesicle trafficking, cell proliferation, differentiation, necrosis, cytoskeleton, and cell migration. It has recently been found that MTMRs are also involved in the occurrence and development of several cardiovascular diseases, including cardiomyocyte hypertrophy, proliferation of vascular smooth muscle cell, LQT1, aortic aneurysm, etc. This review summarizes the functions of MTMRs and highlights their pathophysiological roles in cardiovascular diseases.

3.
Eur J Pharmacol ; 968: 176354, 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38316248

RESUMO

Sepsis is a life-threatening condition involving dysfunctional organ responses stemming from dysregulated host immune reactions to various infections. The lungs are most prone to failure during sepsis, resulting in acute lung injury (ALI). ALI is associated with oxidative stress and inflammation, and current therapeutic strategies are limited. To develop a more specific treatment, this study aimed to synthesise Prussian blue nanozyme (PBzyme), which can reduce oxidative stress and inflammation, to alleviate ALI. PBzyme with good biosafety was synthesised using a modified hydrothermal method. PBzyme was revealed to be an activator of haem oxygenase-1 (HO-1), improving survival rate and ameliorating lung injury in mice. Zinc protoporphyrin, an inhibitor of HO-1, inhibited the prophylactic therapeutic efficacy of PBzyme on ALI, and affected the nuclear factor-κB signaling pathway and activity of HO-1. This study demonstrates that PBzyme can alleviate oxidative stress and inflammation through HO-1 and has a prophylactic therapeutic effect on ALI. This provides a new strategy and direction for the clinical treatment of sepsis-induced ALI.


Assuntos
Lesão Pulmonar Aguda , Ferrocianetos , Sepse , Camundongos , Animais , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/prevenção & controle , Heme Oxigenase-1/metabolismo , Pulmão , Inflamação/complicações , Inflamação/tratamento farmacológico , Sepse/complicações , Sepse/tratamento farmacológico , Lipopolissacarídeos/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo
4.
Atherosclerosis ; 390: 117470, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38342025

RESUMO

BACKGROUND AND AIMS: Myotubularin-related protein 7 (MTMR7) suppresses proliferation in various cell types and is associated with cardiovascular and cerebrovascular diseases. However, whether MTMR7 regulates vascular smooth muscle cell (VSMC) and vascular intimal hyperplasia remains unclear. We explored the role of MTMR7 in phenotypic switching of VSMC and vascular intimal hyperplasia after injury. METHODS AND RESULTS: MTMR7 expression was significantly downregulated in injured arteries. Compared to wild type (WT) mice, Mtmr7-transgenic (Mtmr7-Tg) mice showed reduced intima/media ratio, decreased percentage of Ki-67-positive cells within neointima, and increased Calponin expression in injured artery. In vitro, upregulating MTMR7 by Len-Mtmr7 transfection inhibited platelet derived growth factor (PDGF)-BB-induced proliferation, migration of VSMC and reversed PDGF-BB-induced decrease in expression of Calponin and SM-MHC. Microarray, single cell sequence, and other bioinformatics analysis revealed that MTMR7 is highly related to glucose metabolism and mammalian target of rapamycin complex 1 (mTORC1). Further experiments confirmed that MTMR7 markedly repressed glycolysis and mTORC1 activity in PDGF-BB-challenged VSMC in vitro. Restoring mTORC1 activity abolished MTMR7-mediated suppression of glycolysis, phenotypic shift in VSMC in vitro and protection against vascular intimal hyperplasia in vivo. Furthermore, upregulating MTMR7 in vitro led to dephosphorylation and dissociation of p62 from mTORC1 in VSMC. External expression of p62 in vitro also abrogated the inhibitory effects of MTMR7 on glycolysis and phenotypic switching in PDGF-BB-stimulated VSMC. CONCLUSIONS: Our study demonstrates that MTMR7 inhibits injury-induced vascular intimal hyperplasia and phenotypic switching of VSMC. Mechanistically, the beneficial effects of MTMR7 are conducted via suppressing p62/mTORC1-mediated glycolysis.


Assuntos
Músculo Liso Vascular , Neointima , Camundongos , Animais , Becaplermina/farmacologia , Becaplermina/metabolismo , Proliferação de Células , Músculo Liso Vascular/patologia , Hiperplasia/patologia , Neointima/metabolismo , Camundongos Transgênicos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/farmacologia , Glucose/metabolismo , Miócitos de Músculo Liso/patologia , Movimento Celular , Células Cultivadas , Mamíferos
5.
Med Sci Monit ; 30: e942757, 2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38213018

RESUMO

BACKGROUND Thoracoscopic lobectomy is accompanied by intense trauma and pain due to impaired chest wall integrity. We aimed to introduce a modified ultrasound-guided serratus anterior plane block (MUG-SAPB) for postoperative analgesia in patients who underwent thoracoscopic lobectomy, and to determine whether it could effectively alleviate postoperative pain and improve recovery quality. MATERIAL AND METHODS Overall, 78 patients randomly received either combined MUG-SAPB (0.25% ropivacaine, 10 mg dexamethasone, 40 mL) with patient-controlled intravenous analgesia (PCIA) or received PCIA alone. The primary outcomes were visual analog scale (VAS) scores at rest and during movement at 4, 8, 12, 20, 24, 48, and 72 h postoperatively. The secondary outcomes included use of opioids during surgery, numbers of rescue analgesics (butorphanol), frequency of patient-controlled analgesia (PCA), comfort score within 24 h postoperatively, and postoperative complications within 72 h. RESULTS Compared to the PCIA group, in the MUG-SAPB group, resting VAS scores at 4-24 h (P<0.05) and movement VAS scores at 4-12 h postoperatively (P<0.05) were lower; intraoperative use of sufentanil and frequency of PCA were less, and less rescue analgesia was used (P=0.02, P=0.04 and P=0.03, respectively). Patients in the MUG-SAPB group had faster first mobilization (P=0.04). The MUG-SAPB group had higher comfort scores than the PCIA group (P=0.03). None of the MUG-SAPB patients had any SAPB-related complications. CONCLUSIONS MUG-SAPB effectively relieved postoperative pain, reduced opioid consumption, and accelerated early ambulation in comparison with PCIA alone in patients who underwent thoracoscopic lobectomy.


Assuntos
Bloqueio Nervoso , Humanos , Bloqueio Nervoso/métodos , Manejo da Dor , Analgesia Controlada pelo Paciente , Dor Pós-Operatória/tratamento farmacológico , Dor Pós-Operatória/etiologia , Analgésicos Opioides/uso terapêutico , Ultrassonografia de Intervenção/métodos
6.
Altern Ther Health Med ; 30(2): 25-29, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37856807

RESUMO

Objective: To assess the efficacy of combining esketamine with dexmedetomidine in laparoscopic gallbladder surgery. Methods: We investigated 110 laparoscopic cholecystectomy patients at Jinan Central Hospital, affiliated with Shandong First Medical University, from April 2019 to March 2020. Patients were randomly assigned to the control group (n = 55) or observation group (n = 55). The control group received dexmedetomidine intravenously at 1 µg/kg and a continuous infusion at 0.5 µg•kg-1•h-1. The observation group received esketamine and dexmedetomidine, with intravenous esketamine at 0.4 mg/kg and a continuous infusion at 0.1 mg/(kg•h). We measured heart rate (HR), systolic blood pressure (SBP), and diastolic blood pressure (DBP) at four-time points: before anesthesia (T0), 30 minutes after anesthesia (T1), extubation (T2), and awakening (T3). We also assessed wake time, post-anesthesia care unit (PACU) stay, and Ramasy and visual analogue scale (VAS) scores at 2, 6, 12, and 24 hours post-surgery. Results: At T0, no significant changes occurred in HR, SBP, and DBP in both groups (P > .05). However, at T1 and T2, HR, SBP, and DBP gradually decreased, with the control group exhibiting lower levels than the observation group (P < .05). These levels returned to baseline at T3. PACU residence and wake times showed no significant differences (P > .05). At 2 hours post-operation, Ramasy scores significantly dropped in the observation group versus the control group (P < .05). At 6, 12, and 24 hours post-operation, Ramasy scores exhibited no significant differences (P > .05). Moreover, at 2, 6, and 12 hours post-operation, VAS scores in the observation group were notably lower than in the control group (P < .05). At 24 hours post-operation, VAS scores revealed no significant differences (P > .05). Adverse reactions within 3 days post-operation did not differ significantly between the groups (P > .05). Conclusions: Combining esketamine with dexmedetomidine enhances the quality of laparoscopic cholecystectomy, alleviates postoperative agitation, accelerates cognitive function recovery, reduces cognitive function impairment, and merits clinical consideration.


Assuntos
Dexmedetomidina , Ketamina , Laparoscopia , Humanos , Dexmedetomidina/uso terapêutico , Vesícula Biliar , Ketamina/uso terapêutico
7.
Biochim Biophys Acta Gen Subj ; 1868(1): 130519, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37952564

RESUMO

BACKGROUND: Emerging studies have shown that FAT atypical cadherin 1 (FAT1) and autophagy separately inhibits and promotes acute myeloid leukemia (AML) proliferation. However, it is unknown whether FAT1 were associated with autophagy in regulating AML proliferation. METHODS: AML cell lines, 6-week-old male nude mice and AML patient samples were used in this study. qPCR/Western blot and cell viability/3H-TdR incorporation assays were separately used to detect mRNA/protein levels and cell activity/proliferation. Luciferase reporter assay was used to examine gene promoter activity. Co-IP analysis was used to detect the binding of proteins. RESULTS: In this study, we for the first time demonstrated that FAT1 inhibited AML proliferation by decreasing AML autophagy level. Moreover, FAT1 weakened AML autophagy level via decreasing autophagy related 4B (ATG4B) expression. Mechanistically, we found that FAT1 reduced the phosphorylated and intranuclear SMAD family member 2/3 (smad2/3) protein levels, thus decreasing the activity of ATG4B gene promoter. Furthermore, we found that FAT1 competitively bound to TGF-ßR II which decreased the binding of TGF-ßR II to TGF-ßR I and the subsequent phosphorylation of TGF-ßR I, thus reducing the phosphorylation and intranuclear smad2/3. The experiments in nude mice showed that knockdown of FAT1 promoted AML autophagy and proliferation in vivo. CONCLUSIONS: Collectively, these results revealed that FAT1 downregulates ATG4B expression via inhibiting TGFß-smad2/3 signaling activity, thus decreasing the autophagy level and proliferation activity of AML cells. GENERAL SIGNIFICANCE: Our study suggested that the "FAT1-TGFß-smad2/3-ATG4B-autophagy" pathway may be a novel target for developing new targeted drugs to AML treatment.


Assuntos
Leucemia Mieloide Aguda , Fator de Crescimento Transformador beta , Camundongos , Animais , Humanos , Masculino , Camundongos Nus , Proliferação de Células , Fator de Crescimento Transformador beta/farmacologia , Leucemia Mieloide Aguda/genética , Autofagia , Caderinas , Proteínas Relacionadas à Autofagia/genética , Cisteína Endopeptidases/metabolismo
8.
Clin Exp Hypertens ; 46(1): 2297642, 2024 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-38147409

RESUMO

Pulmonary arterial hypertension (PAH) is characterized by progressive vascular remodeling caused by the excessive proliferation and survival of pulmonary artery smooth muscle cells (PASMCs). Dual-specificity tyrosine regulated kinase 1A (DYRK1A) is a pleiotropic kinase involved in the regulation of multiple biological functions, including cell proliferation and survival. However, the role and underlying mechanisms of DYRK1A in PAH pathogenesis remain unclear. We found that DYRK1A was upregulated in PASMCs in response to hypoxia, both in vivo and in vitro. Inhibition of DYRK1A by harmine significantly attenuated hypoxia-induced pulmonary hypertension and pulmonary artery remodeling. Mechanistically, we found that DYRK1A promoted pulmonary arterial remodeling by enhancing the proliferation and survival of PASMCs through activating the STAT3/Pim-1/NFAT pathway, because STAT3 gain-of-function via adeno-associated virus serotype 2 (AAV2) carrying the constitutively active form of STAT3 (STAT3C) nearly abolished the protective effect of harmine on PAH. Collectively, our results reveal a significant role for DYRK1A in pulmonary arterial remodeling and suggest it as a drug target with translational potential for the treatment of PAH.


Assuntos
Hipertensão Pulmonar , Hipertensão Arterial Pulmonar , Humanos , Hipertensão Arterial Pulmonar/metabolismo , Remodelação Vascular , Harmina/efeitos adversos , Harmina/metabolismo , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/metabolismo , Artéria Pulmonar , Hipóxia , Miócitos de Músculo Liso/metabolismo , Proliferação de Células , Células Cultivadas , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/farmacologia
9.
Neuropsychiatr Dis Treat ; 19: 2573-2583, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38046832

RESUMO

Purpose: We aimed to explore whether anxiety is a risk factor for acute mountain sickness [AMS] in a young Chinese male population. Patients and Methods: A total of 143 young Chinese men with a median age of 23 years (IQR, 21-25) were employed in the present study, and they were divided into the AMS+ and AMS- groups according to the Lake Louise AMS score [AMS-S] after exposure at 3800 m for two days. Participants' pulse oximeter saturation [SpO2] and heart rate [HR] were measured. AMS was evaluated using the AMS-S. The anxiety and sleep quality of the subjects were assessed using the Zung Self-Rating Anxiety Scale [SAS] and the Athens Insomnia Scale [AIS], respectively. Outcomes were analysed using Spearman's partial correlation and logistic regression analysis. Results: After two days of exposure at 3800 m, the overall prevalence of AMS was 54% in the whole group. The HR was significantly higher in the AMS+ group than in the AMS- group, as well as the SAS score and AIS score. A converse pattern was observed for SpO2. A significant difference was observed for the change in SAS and AIS score between the AMS+ and AMS- groups. Correlation analysis showed that AMS-S was positively correlated with SAS score, AIS score, HR, ΔSAS score, ΔAIS score, and ΔHR but negatively correlated with SpO2. AIS score was positively correlated with SAS score. After logistic regression analysis was adjusted for HR, SpO2, ΔAIS and ΔHR, SAS score (OR=1.446, 95% CI 1.200-1.744, p<0.001), AIS score (OR=1.216, 95% CI 1.033-1.432) and ΔSAS score (OR=1.158, 95% CI 1.012-1.327) were identified as independent risk factors for AMS. Conclusion: The present study suggests that anxiety is a risk factor for AMS among young Chinese men, and poor sleep quality may partially mediate the association.

10.
Clin Exp Hypertens ; 45(1): 2278205, 2023 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-37970663

RESUMO

Pulmonary arterial hypertension (PAH) is a disease with a high mortality and few treatment options to prevent the development of pulmonary vessel remodeling, pulmonary vascular resistance, and right ventricular failure. Canagliflozin, a sodium-glucose cotransporter 2 (SGLT2) inhibitor, is originally used in diabetes patients which could assist the glucose excretion and decrease blood glucose. Recently, a few studies have reported the protective effect of SGLT2 inhibitor on monocrotaline-induced PAH. However, the effects of canagliflozin on hypobaric hypoxia-induced PAH as well as its mechanism still unclear. In this study, we used hypobaric hypoxia-induced PAH mice model to demonstrate if canagliflozin could alleviate PAH and prevent pulmonary vessel remodeling. We found that daily canagliflozin administration significantly improved survival in mice with hypobaric hypoxia-induced PAH compared to vehicle control. Canagliflozin treatment significantly reduced right ventricular systolic pressure and increased pulmonary acceleration time determined by hemodynamic assessments. Canagliflozin significantly reduced medial wall thickening and decreased muscularization of pulmonary arterioles compared to vehicle treated mice. In addition, canagliflozin inhibited the proliferation and migration of pulmonary arterial smooth muscle cells through suppressing glycolysis and reactivating AMP-activated protein kinase signaling pathway under hypoxia condition. In summary, our findings suggest that canagliflozin is sufficient to inhibit pulmonary arterial remodeling which is a potential therapeutic strategy for PAH treatment.


Assuntos
Hipertensão Pulmonar , Hipertensão Arterial Pulmonar , Humanos , Camundongos , Animais , Hipertensão Arterial Pulmonar/tratamento farmacológico , Hipertensão Arterial Pulmonar/etiologia , Hipertensão Arterial Pulmonar/metabolismo , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/metabolismo , Canagliflozina/efeitos adversos , Artéria Pulmonar , Hipóxia/complicações , Hipóxia/metabolismo , Miócitos de Músculo Liso/metabolismo , Proliferação de Células , Glucose/farmacologia , Remodelação Vascular , Monocrotalina/farmacologia
11.
Brief Bioinform ; 24(6)2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37798250

RESUMO

Cell-surface proteins play a critical role in cell function and are primary targets for therapeutics. CITE-seq is a single-cell technique that enables simultaneous measurement of gene and surface protein expression. It is powerful but costly and technically challenging. Computational methods have been developed to predict surface protein expression using gene expression information such as from single-cell RNA sequencing (scRNA-seq) data. Existing methods however are computationally demanding and lack the interpretability to reveal underlying biological processes. We propose CrossmodalNet, an interpretable machine learning model, to predict surface protein expression from scRNA-seq data. Our model with a customized adaptive loss accurately predicts surface protein abundances. When samples from multiple time points are given, our model encodes temporal information into an easy-to-interpret time embedding to make prediction in a time-point-specific manner, and is able to uncover noise-free causal gene-protein relationships. Using three publicly available time-resolved CITE-seq data sets, we validate the performance of our model by comparing it with benchmarking methods and evaluate its interpretability. Together, we show that our method accurately and interpretably profiles surface protein expression using scRNA-seq data, thereby expanding the capacity of CITE-seq experiments for investigating molecular mechanisms involving surface proteins.


Assuntos
Algoritmos , Perfilação da Expressão Gênica , Perfilação da Expressão Gênica/métodos , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Proteínas de Membrana
12.
Phytother Res ; 37(12): 5854-5870, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37655750

RESUMO

Doxorubicin (DOX) has aroused contradiction between its potent anti-tumor capacity and severe cardiotoxicity. Galangin (Gal) possesses antioxidant, anti-inflammatory, and antiapoptotic activities. We aimed to explore the role and underlying mechanisms of Gal on DOX-induced cardiotoxicity. Mice were intraperitoneally injected with DOX (3 mg/kg, every 2 days for 2 weeks) to generate cardiotoxicity model and Gal (15 mg/kg, 2 weeks) was co-administered via gavage daily. Nuclear factor erythroid 2-related factor 2 (Nrf2) specific inhibitor, ML385, was employed to explore the underlying mechanisms. Compared to DOX-insulted mice, Gal effectively improved cardiac dysfunction and ameliorated myocardial damage. DOX-induced increase of reactive oxygen species, malondialdehyde, and NADPH oxidase activity and downregulation of superoxide dismutase (SOD) activity were blunted by Gal. Gal also markedly blocked increase of IL-1ß, IL-6, and TNF-α in DOX-insulted heart. Mechanistically, Gal reversed DOX-induced downregulation of Nrf2, HO-1, and promoted nuclear translocation of Nrf2. ML385 markedly blunted the cardioprotective effects of Gal, as well as inhibitive effects on oxidative stress and inflammation. Gal ameliorates DOX-induced cardiotoxicity by suppressing oxidative stress and inflammation via activating Nrf2/HO-1 signaling pathway. Gal may serve as a promising cardioprotective agent for DOX-induced cardiotoxicity.


Assuntos
Cardiotoxicidade , Heme Oxigenase-1 , Camundongos , Animais , Cardiotoxicidade/tratamento farmacológico , Heme Oxigenase-1/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Apoptose , Estresse Oxidativo , Doxorrubicina/efeitos adversos , Transdução de Sinais , Inflamação/metabolismo , Miócitos Cardíacos
13.
Nat Commun ; 14(1): 5148, 2023 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-37620317

RESUMO

Nitrogen-containing heterocycles are the key components in many pharmaceuticals and functional materials. In this study, we report a transition metal-catalyzed high-order reaction sequence for synthesizing a structurally unique N-center 5,6,7-perifused cycle (NCPC). The key characteristics include the formation of a seven-membered ring by the 8π electrocyclization of various alkenes and aromatic heterocycles as π-components, in which metal carbene species are generated that further induce the cleavage of the α-C-H or -C-C bond. Specifically, the latter can react with various nucleophilic reagents containing -O, -S, -N, and -C. The stereo-controlled late-stage modification of some complicated pharmaceuticals indicates the versatility of this protocol.

14.
J Phys Chem B ; 127(28): 6374-6384, 2023 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-37418387

RESUMO

Aqueous stability is a critical property for the application of metal-organic framework (MOF) materials in humid conditions. The sampling of the free energy surface for a water reaction is challenging due to a lack of a reactive force field. Here, we developed a ReaxFF force field for simulating the reaction of zeolitic imidazole frameworks (ZIFs) with water. We carried out metadynamics simulations based on ReaxFF to study the reaction of water with a few different types of MOFs. We also conducted an experimental water immersion test and characterized the XRD, TG, and gas adsorption properties of the MOFs before and after the immersion test. By considering the energy barrier for a hydrolysis reaction, the simulation results are in good agreement with the experiments. MOFs with open structures and large pores are found to be unstable in metadynamics simulations, where the water molecule can attack or bond with the metallic node relatively easily. In contrast, it is more difficult for water to attack the Zn atom in the ZnN4 tetrahedral structure of ZIFs. We also found that ZIFs with the -NO2 functional groups have higher water stability. Discrepancies between the metadynamics simulation and gas adsorption experiments have been accounted for from the phase/crystallinity change of the structure reflected in the X-ray diffraction and thermogravimetry analysis of the MOF samples.

15.
Phytomedicine ; 117: 154922, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37321078

RESUMO

BACKGROUND: Doxorubicin (DOX) is a potent anticancer chemotherapeutic agent whose clinical application is substantially constrained by its cardiotoxicity. The pathophysiology of DOX-induced cardiotoxicity manifests as cardiomyocyte pyroptosis and inflammation. Amentoflavone (AMF) is a naturally occurring biflavone possessing anti-pyroptotic and anti-inflammatory properties. However, the mechanism through which AMF alleviates DOX-induced cardiotoxicity remains undetermined. PURPOSE: This study aimed at investigating the role of AMF in alleviating DOX-induced cardiotoxicity. STUDY DESIGN AND METHODS: To assess the in vivo effect of AMF, DOX was intraperitoneally administered into a mouse model to induce cardiotoxicity. To elucidate the underlying mechanisms, the activities of STING/NLRP3 were quantified using the NLRP3 agonist nigericin and the STING agonist amidobenzimidazole (ABZI). Primary cardiomyocytes isolated from neonatal Sprague-Dawley rats were treated with saline (vehicle) or DOX with or without AMF and/or ABZI. The echocardiogram, haemodynamics, cardiac injury markers, heart/body weight ratio, and pathological alterations were monitored; the STING/NLRP3 pathway-associated proteins were detected by western blot and cardiomyocyte pyroptosis was analysed by immunofluorescence staining of cleaved N-terminal GSDMD and scanning electron microscopy. Furthermore, we evaluated the potential of AMF in compromising the anticancer effects of DOX in human breast cancer cell lines. RESULTS: AMF substantially alleviated cardiac dysfunction and reduced heart/body weight ratio and myocardial damage in mice models of DOX-induced cardiotoxicity. AMF effectively suppressed DOX-mediated upregulation of IL-1ß, IL-18, TNF-α, and pyroptosis-related proteins, including NLRP3, cleaved caspase-1, and cleaved N-terminal GSDMD. The levels of apoptosis-related proteins, namely Bax, cleaved caspase-3, and BCL-2 were not affected. In addition, AMF inhibited STING phosphorylation in DOX-affected hearts. Intriguingly, the administration of nigericin or ABZI dampened the cardioprotective effects of AMF. The in vitro anti-pyroptotic effect of AMF was demonstrated in attenuating the DOX-induced reduction in cardiomyocyte cell viability, upregulation of cleaved N-terminal GSDMD, and pyroptotic morphology alteration at the microstructural level. AMF exhibited a synergistic effect with DOX to reduce the viability of human breast cancer cells. CONCLUSION: AMF alleviates DOX-induced cardiotoxicity by suppressing cardiomyocyte pyroptosis and inflammation via inhibition of the STING/NLRP3 signalling pathway, thereby validating its efficacy as a cardioprotective agent.


Assuntos
Neoplasias da Mama , Miócitos Cardíacos , Ratos , Camundongos , Animais , Humanos , Feminino , Piroptose , Cardiotoxicidade/tratamento farmacológico , Cardiotoxicidade/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Nigericina/efeitos adversos , Nigericina/metabolismo , Ratos Sprague-Dawley , Doxorrubicina/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo , Inflamação/metabolismo , Neoplasias da Mama/patologia , Peso Corporal
16.
J Cardiovasc Pharmacol ; 82(3): 221-228, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37381169

RESUMO

ABSTRACT: Excessive proliferation and migration of vascular smooth muscle cells (VSMCs) cause neointimal hyperplasia after percutaneous vascular interventions. Nuclear receptor subfamily 1 group D member 1 (NR1D1), a crucial member of circadian clock, is involved in the regulation of atherosclerosis and cellular proliferation. However, whether NR1D1 affects vascular neointimal hyperplasia remains unclear. In this study, we found that activating NR1D1 reduced injury-induced vascular neointimal hyperplasia. Overexpression of NR1D1 reduced the number of Ki-67-positive VSMCs and migrated VSMCs after platelet-derived growth factor (PDGF)-BB treatment. Mechanistically, NR1D1 suppressed the phosphorylation of AKT and 2 main effectors of the mammalian target of rapamycin complex 1 (mTORC1), S6, and 4EBP1 in PDGF-BB-challenged VSMCs. Re-activation of mTORC1 by Tuberous sclerosis 1 siRNA (si Tsc1 ) and re-activation of AKT by SC-79 abolished NR1D1-mediated inhibitory effects on proliferation and migration of VSMCs. Moreover, decreased mTORC1 activity induced by NR1D1 was also reversed by SC-79. Simultaneously, Tsc1 knockdown abolished the vascular protective effects of NR1D1 in vivo. In conclusion, NR1D1 reduces vascular neointimal hyperplasia by suppressing proliferation and migration of VSMCs in an AKT/mTORC1-dependent manner.


Assuntos
Músculo Liso Vascular , Lesões do Sistema Vascular , Humanos , Hiperplasia/metabolismo , Músculo Liso Vascular/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proliferação de Células , Becaplermina/farmacologia , Lesões do Sistema Vascular/patologia , Neointima/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Miócitos de Músculo Liso/metabolismo , Movimento Celular , Células Cultivadas
17.
Nucleic Acids Res ; 51(13): 6578-6592, 2023 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-37246643

RESUMO

In this paper, we introduce Gene Knockout Inference (GenKI), a virtual knockout (KO) tool for gene function prediction using single-cell RNA sequencing (scRNA-seq) data in the absence of KO samples when only wild-type (WT) samples are available. Without using any information from real KO samples, GenKI is designed to capture shifting patterns in gene regulation caused by the KO perturbation in an unsupervised manner and provide a robust and scalable framework for gene function studies. To achieve this goal, GenKI adapts a variational graph autoencoder (VGAE) model to learn latent representations of genes and interactions between genes from the input WT scRNA-seq data and a derived single-cell gene regulatory network (scGRN). The virtual KO data is then generated by computationally removing all edges of the KO gene-the gene to be knocked out for functional study-from the scGRN. The differences between WT and virtual KO data are discerned by using their corresponding latent parameters derived from the trained VGAE model. Our simulations show that GenKI accurately approximates the perturbation profiles upon gene KO and outperforms the state-of-the-art under a series of evaluation conditions. Using publicly available scRNA-seq data sets, we demonstrate that GenKI recapitulates discoveries of real-animal KO experiments and accurately predicts cell type-specific functions of KO genes. Thus, GenKI provides an in-silico alternative to KO experiments that may partially replace the need for genetically modified animals or other genetically perturbed systems.


Assuntos
Redes Reguladoras de Genes , Análise de Célula Única , Animais , Técnicas de Inativação de Genes , Regulação da Expressão Gênica , Análise de Sequência de RNA , Perfilação da Expressão Gênica
18.
Brain Behav ; 13(6): e2998, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37095720

RESUMO

OBJECTIVE: Cerebral venous sinus thrombosis (CVST) is believed to be associated with high-altitude exposure and has worse clinical prognosis in plateau areas than in plain areas, although this needs to be further verified. This retrospective study aims to compare the clinical differences of patients with CVST in plateau and plain areas and further ascertain the role of high-altitude exposure in the etiology of aggravating predisposition toward CVST. METHODS: Twenty-four symptomatic CVST patients occurring at plateau areas (altitude ≥ 4000 m), in corresponding with 24 CVST patients occurring at plain areas (altitude ≤ 1000 m), were recruited according to the inclusion and exclusion criteria from June 2020 to December 2021. The collected data and compared parameters include clinical features, neuroimaging findings, hematology profile, lipid profile, and coagulation profile within 24 h of hospital admission, as well as the treatment method and final outcome. RESULTS: There were no obvious differences of demographic characteristics, including gender, age, height, and weight between patients with CVST in plateau and plain areas, as well as medical history, neuroimaging findings, treatment protocols, and clinical outcome (all p > .05). Compared to patients with CVST at plain areas, time before hospital admission was longer and heartbeat was slower in patients with CVST at plateau areas (all p < .05). More importantly, elevated red blood cells counts, hemoglobin level, and altered coagulation function were found in patients with CVST at plateau areas (all p < .05). CONCLUSION: CVST patients in plateau areas presented with altered clinical characteristics, altered coagulation function, and aggravated predisposition toward venous thromboembolism compared with CVST patients in plain areas. Future prospective studies will be needed to further elucidate the influences of a high altitude on the pathogenesis of CVST.


Assuntos
Trombose dos Seios Intracranianos , Humanos , Estudos Retrospectivos , Estudos Prospectivos , Trombose dos Seios Intracranianos/diagnóstico por imagem , Trombose dos Seios Intracranianos/etiologia , Trombose dos Seios Intracranianos/tratamento farmacológico , Prognóstico , Neuroimagem
19.
Neuroimmunomodulation ; 30(1): 55-68, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36858024

RESUMO

INTRODUCTION: Neuroinflammation contributes to secondary injury after traumatic brain injury (TBI), which has been mainly mediated by the microglia. MiR-124 was reported to play an important role in the polarization of microglia by targeting TLR4 signaling pathway. However, the role and mechanism of miR-124 in neuroinflammation mediated by microglia after TBI is unclear. To clarify this, we performed this research. METHODS: The expression of miR-124 was first measured by RT-PCR in the injured brain at 1/3/7 days post-TBI. Then, miR-124 mimics or inhibitors administration was used to interfere the expression of miR-124 at 24 h post-TBI. Subsequently, the microglia polarization markers were detected by RT-PCR, the expression of inflammatory cytokines was detected by ELISA, the expression of TLR4/MyD88/IRAK1/TRAF6/NF-κB was measured by WB, and the neurological deficit was evaluated by NSS and MWM test. At last, in vitro experiments were performed to explore the exact target molecule of miR-124 on TLR4 signaling pathway. RESULTS: Animal research indicated that the expression of miR-124 was downregulated after TBI. Upregulation of miR-124 promoted the M2 polarization of microglia and inhibited the activity of TLR4 pathway, as well as reduced neuroinflammation and neurological deficit after TBI. In vitro experiments indicated that miR-124 promoted the M2 polarization of microglia and reduced neuroinflammation by inhibiting TRAF6. CONCLUSION: This study demonstrated that upregulation of miR-124 promoted the M2 polarization of microglia and reduced neuroinflammation after TBI by inhibiting TRAF6.


Assuntos
Lesões Encefálicas Traumáticas , MicroRNAs , Animais , Fator 6 Associado a Receptor de TNF/metabolismo , Doenças Neuroinflamatórias , Receptor 4 Toll-Like , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/metabolismo , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Microglia/metabolismo
20.
Clin Exp Hypertens ; 45(1): 2178659, 2023 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-36794491

RESUMO

BACKGROUND: In-stent restenosis hardly limits the therapeutic effect of the percutaneous vascular intervention. Although the restenosis is significantly ameliorated after the application of new drug-eluting stents, the incidence of restenosis remains at a high level. OBJECTIVE: Vascular adventitial fibroblasts (AFs) play an important role in intimal hyperplasia and subsequent restenosis. The current study was aimed to investigate the role of nuclear receptor subfamily 1, group D, member 1 (NR1D1) in the vascular intimal hyperplasia. METHODS AND RESULTS: We observed increased expression of NR1D1 after the transduction of adenovirus carrying Nr1d1 gene (Ad-Nr1d1) in AFs. Ad-Nr1d1 transduction significantly reduced the numbers of total AFs, Ki-67-positive AFs, and the migration rate of AFs. NR1D1 overexpression decreased the expression level of ß-catenin and attenuated the phosphorylation of the effectors of mammalian target of rapamycin complex 1 (mTORC1), including mammalian target of rapamycin (mTOR) and 4E binding protein 1 (4EBP1). Restoration of ß-catenin by SKL2001 abolished the inhibitory effects of NR1D1 overexpression on the proliferation and migration of AFs. Surprisingly, the restoration of mTORC1 activity by insulin could also reverse the decreased expression of ß-catenin, attenuated proliferation, and migration in AFs induced by NR1D1 overexpression. In vivo, we found that SR9009 (an agonist of NR1D1) ameliorated the intimal hyperplasia at days 28 after injury of carotid artery. We further observed that SR9009 attenuated the increased Ki-67-positive AFs, an essential part of vascular restenosis at days 7 after injury to the carotid artery. CONCLUSION: These data suggest that NR1D1 inhibits intimal hyperplasia by suppressing the proliferation and migration of AFs in a mTORC1/ß-catenin-dependent manner.


Assuntos
Músculo Liso Vascular , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares , beta Catenina , beta Catenina/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Fibroblastos , Hiperplasia/metabolismo , Hiperplasia/patologia , Antígeno Ki-67/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Miócitos de Músculo Liso , Neointima/genética , Neointima/metabolismo , Neointima/patologia , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA