Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
BMC Genomics ; 25(1): 254, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38448814

RESUMO

BACKGROUND: Neddylation, an important post-translational modification (PTM) of proteins, plays a crucial role in follicular development. MLN4924 is a small-molecule inhibitor of the neddylation-activating enzyme (NAE) that regulates various biological processes. However, the regulatory mechanisms of neddylation in rabbit ovarian cells have not been emphasized. Here, the transcriptome and metabolome profiles in granulosa cells (GCs) treated with MLN4924 were utilized to identify differentially expressed genes, followed by pathway analysis to precisely define the altered metabolisms. RESULTS: The results showed that 563 upregulated and 910 downregulated differentially expressed genes (DEGs) were mainly enriched in pathways related to cancer, cell cycle, PI3K-AKT, progesterone-mediated oocyte maturation, and PPAR signaling pathway. Furthermore, we characterized that MLN4924 inhibits PPAR-mediated lipid metabolism, and disrupts the cell cycle by promoting the apoptosis and proliferation of GCs. Importantly, we found the reduction of several metabolites in the MLN4924 treated GCs, including glycerophosphocholine, arachidic acid, and palmitic acid, which was consistent with the deregulation of PPAR signaling pathways. Furthermore, the increased metabolites included 6-Deoxy-6-sulfo-D-glucono-1,5-lactone and N-Acetyl-D-glucosaminyldiphosphodolichol. Combined with transcriptome data analyses, we identified genes that strongly correlate with metabolic dysregulation, particularly those related to glucose and lipid metabolism. Therefore, neddylation inhibition may disrupt the energy metabolism of GCs. CONCLUSIONS: These results provide a foundation for in-depth research into the role and molecular mechanism of neddylation in ovary development.


Assuntos
Ciclopentanos , Receptores Ativados por Proliferador de Peroxissomo , Fosfatidilinositol 3-Quinases , Pirimidinas , Feminino , Animais , Coelhos , Células da Granulosa , Metabolismo dos Lipídeos
2.
PeerJ ; 12: e16748, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38304189

RESUMO

Acute lung injury (ALI) is one of the most serious complications of sepsis, characterized by high morbidity and mortality rates. Ferroptosis has recently been reported to play an essential role in sepsis-induced ALI. Excessive neutrophil extracellular traps (NETs) formation induces exacerbated inflammation and is crucial to the development of ALI. In this study, we explored the effects of ferroptosis and NETs and observed the therapeutic function of mesenchymal stem cells (MSCs) on sepsis-induced ALI. First, we produced a cecal ligation and puncture (CLP) model of sepsis in rats. Ferrostain-1 and DNase-1 were used to inhibit ferroptosis and NETs formation separately, to confirm their effects on sepsis-induced ALI. Next, U0126 was applied to suppress the MEK/ERK signaling pathway, which is considered to be vital to NETs formation. Finally, the therapeutic effect of MSCs was observed on CLP models. The results demonstrated that both ferrostain-1 and DNase-1 application could improve sepsis-induced ALI. DNase-1 inhibited ferroptosis significantly in lung tissues, showing that ferroptosis could be regulated by NETs formation. With the inhibition of the MEK/ERK signaling pathway by U0126, NETs formation and ferroptosis in lung tissues were both reduced, and sepsis-induced ALI was improved. MSCs also had a similar protective effect against sepsis-induced ALI, not only inhibiting MEK/ERK signaling pathway-mediated NETs formation, but also alleviating ferroptosis in lung tissues. We concluded that MSCs could protect against sepsis-induced ALI by suppressing NETs formation and ferroptosis in lung tissues. In this study, we found that NETs formation and ferroptosis were both potential therapeutic targets for the treatment of sepsis-induced ALI, and provided new evidence supporting the clinical application of MSCs in sepsis-induced ALI treatment.


Assuntos
Lesão Pulmonar Aguda , Butadienos , Armadilhas Extracelulares , Ferroptose , Células-Tronco Mesenquimais , Nitrilas , Sepse , Ratos , Animais , Armadilhas Extracelulares/metabolismo , Lesão Pulmonar Aguda/etiologia , Desoxirribonuclease I/farmacologia , Sepse/complicações , Células-Tronco Mesenquimais/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/efeitos adversos
3.
Zhongguo Fei Ai Za Zhi ; 26(9): 639-649, 2023 Sep 20.
Artigo em Chinês | MEDLINE | ID: mdl-37985150

RESUMO

BACKGROUND: Non-small cell lung cancer (NSCLC) is one of the most lethal malignancies worldwide. A novel Chinese medicine formula-01 (NCHF-01) has shown significant clinical efficacy in the treatment of NSCLC, but the mechanism of this formula in the treatment of NSCLC is not fully understood. The aim of this study is to investigate the molecular mechanism of NCHF-01 in inhibiting NSCLC. METHODS: Lewis lung cells (LLC) tumor bearing mice were established to detect the tumor inhibitory effect of NCHF-01. The morphological changes of tissues and organs in LLC tumor-bearing mice were detected by hematoxylin-eosin (HE) staining. NSCLC cells were treated by NCHF-01. The effects of cell viability and proliferation were detected by MTT and crystal violet staining experiment. Flow cytometry was used to detect cell cycle, apoptosis and reactive oxygen species (ROS). Network pharmacology was used to predict the mechanism of its inhibitory effect of NSCLC. Western blot and immunohistochemistry (IHC) were used to detect the expression of related proteins. RESULTS: NCHF-01 can inhibit tumor growth in LLC tumor-bearing mice, and has no obvious side effects on other tissues and organs. NCHF-01 could inhibit cell viability and proliferation, induce G2/M phase arrest and apoptosis, and promote the increase of ROS level. Network pharmacological analysis showed that NCHF-01 exerts anti-NSCLC effects through various biological processes such as oxidative stress and central carbon metabolism. NCHF-01 can reduce the protein expression and enzyme activity of the key enzymes 6-phosphate glucose dehydrogenase (G6PD) and 6-phosphogluconate dehydrogenase (6PGD) in the pentose phosphate pathway (PPP). CONCLUSIONS: NCHF-01 can inhibit NSCLC through oxidative stress dependent on the PPP.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Animais , Camundongos , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/farmacologia , Espécies Reativas de Oxigênio/uso terapêutico , Medicina Tradicional Chinesa , Via de Pentose Fosfato , Estresse Oxidativo , Linhagem Celular Tumoral , Proliferação de Células , Apoptose
4.
J Med Microbiol ; 72(10)2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37855710

RESUMO

Introduction. Oxymatrine is a natural quinazine alkaloid extracted from Sophora flavescens and has many medicinal values. Oxymatrine showed protective effects, viral inhibition and effects against lung cancer.Hypothesis/Gap Statement. Individuals with lung cancer exhibit heightened vulnerability to COVID-19 infection due to compromised immune function. In conjunction with COVID-19, it is hypothesized that oxymatrine may exert potent pharmacological effects on lung cancer patients.Aim. The objective of this study was to assess the pharmacological mechanisms and targets of oxymatrine in relation to COVID-19 lung cancer.Methodology. Utilizing network pharmacology analysis, a selection of 2628 genes were identified as co-targets for both COVID-19 and lung cancer. Subsequently, a clinicopathological analysis was conducted by integrating RNA-Seq and clinical data obtained from the TCGA-LUAD lung cancer dataset, which was acquired from the official TCGA website. The identification of pharmacological targets for oxymatrine was accomplished through the utilization of various databases including Pharm mapper, SWISS Target prediction, and STITCH. These identified targets were further investigated for protein-protein interaction (PPI) using STRING, as well as for gene ontology (GO) and KEGG pathways.Results. The effects of oxymatrine on COVID-19-induced lung cancer were mediated by immune regulation, cytoprotection, antiviral, and anti-inflammatory activities, immune regulation, and control of related signalling pathways, including the formation of the neutrophil extracellular trap, phagosome, Toll-like receptor signalling pathway, apoptosis, proteoglycans in cancer, extracellular matrix disassembly, and proteolysis involved in cellular protein catabolism. Furthermore, important substances and genes like ALB, MMP3, MMP1, and TLR4 may affect how oxymatrine suppresses lung cancer/COVID-19 development.Conclusion. To treat COVID-19 or lung cancer paired with COVID-19, oxymatrine may improve the therapeutic efficacy of current clinical antiviral medicines and immunotherapy.


Assuntos
Alcaloides , COVID-19 , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Farmacologia em Rede , Alcaloides/farmacologia , Alcaloides/uso terapêutico , Antivirais/farmacologia , Antivirais/uso terapêutico
5.
Transl Res ; 262: 44-59, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37507007

RESUMO

Liver transplantation is the ultimate treatment option for end-stage liver failure. However, liver graft injury remains a challenge. This study aimed to investigate the role of connexin32 (Cx32) in liver graft injury and elucidate its mechanism of action. Through detecting liver graft samples from 6 patients, we observed that changes in the Cx32 level coincided with liver graft injury. Therefore, we established autologous orthotopic liver transplantation (AOLT) models using Cx32-knockout and wild-type mice and hypoxia/reoxygenation (H/R) and lipopolysaccharide (LPS) pretreatment models using alpha mouse liver 12 (AML12) cells, to explore Cx32 mechanisms in liver graft injury. Following in vivo and in vitro Cx32 knockout, oxidative stress and inflammatory response were inhibited through the regulation of PKC-α/NF-κB/NLRP3 and Nrf2/NOX4/ROS signaling pathways, thereby reducing Bak/Bax-related apoptosis and ameliorating liver graft injury. When the Cx32-based gap junction (GJ) was blocked with 2-aminoethoxydiphenyl borate (2-APB), ROS transfer was attenuated between neighboring cells, exacerbated oxidative stress and inflammatory response were prevented, and aggravation of liver graft injury was mitigated. These results highlight the dual regulation mechanism of Cx32 in liver graft injury. Through interaction with PKC-α, Cx32 regulated the NF-κB/NLRP3 and Nrf2/NOX4/ROS signaling pathways, thus directly triggering oxidative stress and inflammatory response. Simultaneously, mass-produced ROS were transferred to neighboring cells through Cx32 channels, for which oxidative stress and the inflammatory response were aggravated indirectly. Finally, Bak/Bax-related apoptosis was activated, thereby worsening liver graft injury. Our findings propose Cx32 as a dual mechanistic factor for oxidative stress and inflammatory signaling pathways in regulating cell apoptosis on liver graft injury, which suggests a promising therapeutic targets for liver graft injury.


Assuntos
Transplante de Fígado , Humanos , Camundongos , Animais , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fator 2 Relacionado a NF-E2 , Proteína X Associada a bcl-2/metabolismo , Fígado/metabolismo , Hepatócitos
6.
Comput Math Methods Med ; 2023: 7550317, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36873790

RESUMO

Background: Clinically, there were few reports on single-hole thoracoscopic segmental resection in non-small-cell lung cancer (NSCLC), and no report on the comparison of single-hole and three-hole thoracoscopic segmental resection. Hence, the purpose of the study was to explore the perioperative role of single-port thoracoscopic segmentectomy and three-port thoracoscopic segmentectomy for early-stage NSCLC. Methods: The clinical data of 80 patients with early-stage NSCLC who were treated in our hospital from January 2021 to June 2022 were selected as the retrospective research subjects and divided into a comparison/research group with 40 cases in each group according to different surgical methods. Among them, the comparison group was received three-port thoracoscopic segmentectomy, and the research group was received single-port thoracoscopic segmentectomy. The surgical indicators, immune and tumor marker levels, as well as prognostic complications between two groups were compared. Results: There was no remarkable diversity between the two groups in terms of operation time and the number of lymph nodes dissected during the operation (P > 0.05). The surgical blood loss in research group was lower than comparison group (P < 0.05). After treatment, the levels of CYFRA21-1, CA125, as well as VGEF in the research group were markedly lower than comparison group (P < 0.05). The differences in CD4+, CD3+, and CD4+/CD8+ after treatment were prominent, and the research group was higher than comparison group (P < 0.05). There was no statistical difference in postoperative complications between the two groups (P > 0.05). Conclusions: Single-hole thoracoscopic lobectomy has obvious advantages in the treatment of NSCLC, which can reduce intraoperative bleeding, enhance the recovery of patients' immune function, and promote postoperative recovery.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Carcinoma de Pequenas Células do Pulmão , Humanos , Pneumonectomia , Estudos Retrospectivos
7.
Cell Prolif ; 56(10): e13461, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36974350

RESUMO

Bone marrow-derived mesenchymal stem cells (BMSCs) show a good property for pain treatment by modulating inflammatory response. However, the underlying therapeutic effect and related mechanism of BMSCs on inflammatory pain remain unclear. Therefore, we explored the function and potential mechanism of BMSCs performing in a complete Freund's adjuvant (CFA)-induced inflammatory pain model in this study. Here, BMSCs were injected into the CFA-treated rats, and we used behavioural tests to evaluate the changes in hypersensitivity. High-throughput sequencing was used to screen out the hub genes. Molecular biology experiments were performed to detect the level of P2X3 or inflammatory mediators in rats and observed the distribution of P2X3 in neural cells. Furthermore, the function of the P2X3 was explored via inhibitor and activator experiments. Finally, we found that BMSCs alleviated hyperalgesia and spinal levels of pro-inflammatory factors in CFA-treated rats. High-throughput sequencing showed that P2X3 and P2X7 were identified as hub genes, and only the expression level of P2X3 was significantly down-regulated after BMSCs treatment. Immunohistochemistry showed that P2X3 mainly colocalized with microglia and astrocytes. The levels of P2X3 and pro-inflammatory factors were all significantly reduced after BMSC injection. Moreover, similar attenuation was found in the CFA-treated rats after injecting the P2X3 inhibitor, and a P2X3 antagonist reversed the attenuation induced by the BMSCs. These findings suggest that BMSCs exerted a therapeutic effect on inflammatory pain by inhibiting the expression of P2X3 and the excessive production of inflammatory mediators was associated with an increased P2X3 level and BMSC therapy reverse these effects.


Assuntos
Medula Óssea , Células-Tronco Mesenquimais , Ratos , Animais , Adjuvante de Freund/efeitos adversos , Medula Óssea/metabolismo , Ratos Sprague-Dawley , Inflamação/induzido quimicamente , Inflamação/terapia , Dor/induzido quimicamente , Dor/complicações , Dor/metabolismo , Células-Tronco Mesenquimais/metabolismo , Mediadores da Inflamação
8.
Clin Pharmacokinet ; 62(3): 435-447, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36735213

RESUMO

BACKGROUND: Data available for pharmacokinetics (PK)/pharmacodynamics (PD) of ticagrelor and significant endogenous/exogenous factors or biomarkers related to bleeding events in both healthy and clinical patients are limited. OBJECTIVE: Based on PK and PD data from multicenter healthy subjects and patients, we aimed to establish an integrated approach towards population PK (pop PK) and the PD model of ticagrelor. METHODS: This study was conducted as a multicenter, prospective clinical registration study involving both healthy subjects and clinical patients. The integrated Pharmacokinetic/pharmacodynamic (PK/PD) models were characterized based on PK/PD [ticagrelor concentration, aggregation baseline (BASE), P2Y12 response unit (PRU) and inhibition rate (INHIBIT)] data from 175 healthy volunteers. The model was corrected by sparse PD (BASE, PRU and INHIBIT) data from 208 patients with acute coronary syndrome (ACS). The correlations between PD biomarkers and clinically relevant bleedings in 1 year were explored. RESULTS: A one-compartment, linear model with first-order absorption was adopted as PK model. Food status (FOOD) and body weight (WT) significantly influenced clearance and improved the fitting degree of the PK model, while SEX was selected as the covariates of the PD model. For patients taking ticagrelor 90 mg, the peak value [mean (95% CI)] of PRU was 355.15 (344.24-366.06) and the trough value was 3.64 (3.14-4.15). The PRU mean parameters were basically within the expected range (80-200) of the literature suggestions. CONCLUSION: A fixed dose of ticagrelor, without adjusting the dosing regimen other than covariates of FOOD/WT/SEX, could be used in patients with acute coronary syndromes, and the standard regimen could be used in Chinese patients from the perspective of exposure.


Assuntos
Síndrome Coronariana Aguda , Inibidores da Agregação Plaquetária , Humanos , Ticagrelor , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Síndrome Coronariana Aguda/tratamento farmacológico , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Estudos Prospectivos , Adenosina
9.
Sheng Li Xue Bao ; 74(5): 843-855, 2022 Oct 25.
Artigo em Chinês | MEDLINE | ID: mdl-36319107

RESUMO

The Forkhead box class O proteins (FOXOs) family consists of highly conserved transcription factors, including FOXO1, FOXO3, FOXO4 and FOXO6. Each member of the FOXOs family is ubiquitously expressed and involved in regulating many biological activities such as tumor cell proliferation, apoptosis, migration and oxidative stress. The activity of FOXOs is mainly regulated by post-translational modification, and its inactivation is mainly mediated by the over-activation of its upstream modifying enzymes, which provides a possibility to use drugs to recover its activity. It is worth noting that FOXOs can not only inhibit, but also promote the occurrence and development of human tumors due to the complex effects of FOXOs. This review will summarize the structure and activity regulation of FOXOs, and discuss their tumor inhibiting effects by limiting cell proliferation and inducing apoptosis, as well as their tumor promoting effects by maintaining cell homeostasis, promoting metastasis and inducing drug resistance, so as to provide new ideas for the pathological research of related diseases and open up new ways to promote broader prevention and treatment strategies.


Assuntos
Neoplasias , Humanos , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Processamento de Proteína Pós-Traducional , Estresse Oxidativo , Apoptose
10.
Mol Biol Rep ; 49(10): 9877-9891, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35553342

RESUMO

Since generally confronting with the hypoxic and stressful microenvironment, cancer cells alter their glucose metabolism pattern to glycolysis to sustain the continuous proliferation and vigorous biological activities. Bifunctional 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFK-2/FBPase-2) isoform 3 (PFKFB3) functions as an effectively modulator of glycolysis and also participates in regulating angiogenesis, cell death and cell stemness. Meanwhile, PFKFB3 is highly expressed in a variety of cancer cells, and can be activated by several regulatory factors, such as hypoxia, inflammation and cellular signals. In colorectal cancer (CRC) cells, PFKFB3 not only has the property of high expression, but also probably relate to inflammation-cancer transformation. Recent studies indicate that PFKFB3 is involved in chemoradiotherapy resistance as well, such as breast cancer, endometrial cancer and CRC. Cancer stem cells (CSCs) are self-renewable cell types that contribute to oncogenesis, metastasis and relapse. Several studies indicate that CSCs utilize glycolysis to fulfill their energetic and biosynthetic demands in order to maintain rapid proliferation and adapt to the tumor microenvironment changes. In addition, elevated PFKFB3 has been reported to correlate with self-renewal and metastatic outgrowth in numerous kinds of CSCs. This review summarizes our current understanding of PFKFB3 roles in modulating cancer metabolism to maintain cell proliferation and stemness, and discusses its feasibility as a potential target for the discovery of antineoplastic agents, especially in CRC.


Assuntos
Antineoplásicos , Neoplasias Colorretais , Proliferação de Células , Transformação Celular Neoplásica , Neoplasias Colorretais/genética , Glucose/metabolismo , Glicólise , Humanos , Inflamação , Recidiva Local de Neoplasia , Células-Tronco Neoplásicas , Fosfofrutoquinase-2/genética , Microambiente Tumoral
11.
Med Oncol ; 39(1): 10, 2021 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-34761330

RESUMO

Numerous studies have shown that 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoform 3 (PFKFB3), a pivotal enzyme in modulating glycolysis, plays vital roles in various physiological processes. PFKFB3 activity could be regulated by several factors, such as hypoxia and AMPK signaling; however, it could also function as upstream of AMPK signaling. Here, we showed that PFKFB3 inhibitor PFK-15 induced cell viability loss and apoptosis. Deprivation of PFKFB3 inhibited autophagy, while enhanced the ubiquitin-proteasome degradation pathway. Furthermore, PFK-15 reduced both the AMPK and AKT-mTORC1 signaling pathways, as the attenuated phosphorylation level of kinases themselves and their substrates. The addition of AICAR rescued the AMPK activity and autophagy, but enhanced PFK-15-induced cell viability loss. In fact, AICAR promoted the cytotoxicity of PFK-15 even in the AMPKα1/2-silenced cells, indicating AICAR might function in an AMPK-independent manner. Nevertheless, AICAR further reduced the AKT-mTORC1 activity down-regulated by PFK-15. Moreover, it failed to enhance PFK-15's cytotoxicity in the AKT1/2-silenced cells, indicating AKT-mTORC1 participated during these processes. Collectively, the presented data demonstrated that PFK-15 inhibited cell viability, AMPK, and AKT-mTORC1 signaling, and AICAR probably enhanced the cell viability loss aroused by PFK-15 in an AKT-dependent and AMPK-independent manner, thereby revealing a more intimate relationship among PFKFB3, AMPK, and AKT-mTORC1 signaling pathways.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Fosfofrutoquinase-2/antagonistas & inibidores , Ribonucleotídeos/farmacologia , Aminoimidazol Carboxamida/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Transdução de Sinais/efeitos dos fármacos
12.
BMC Anesthesiol ; 21(1): 267, 2021 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-34732147

RESUMO

BACKGROUND: Opioids have been identified by the World Health Organization to be 'indispensable for the relief of pain and suffering'. Side-effects, such as nausea, vomiting, postoperative delirium, and effects on breathing, of opioids have been well investigated; however, the influence of opioids on monocyte-endothelial adherence has never been reported. Therefore, we explored the effects of representative opioids, fentanyl, sufentanil, and remifentanil, on monocyte-endothelial adherence and the underlying mechanisms. METHODS: We built a cell adhesion model with U937 monocytes and human umbilical vein endothelial cells (HUVECs). Two kinds of connexin43 (Cx43) channel inhibitors, 18-α-GA and Gap 27, were used to alter Cx43 channel function in U937 monocytes and HUVECs, respectively, to determine the effects of Cx43 channels on U937-HUVEC adhesion. Subsequently, the effects of fentanyl, sufentanil and remifentanil on Cx43 channel function and U937-HUVEC adhesion were explored. RESULTS: When fentanyl, sufentanil and remifentanil acted on monocytes or endothelial cells, their effects on monocyte-endothelial adherence differed. When acting on U937 monocytes, sufentanil significantly increased U937-HUVEC adhesion which was associated with reduced release of ATP from Cx43 channels, while fentanyl and remifentanil did not have these influences. Although sufentanil could also inhibit Cx43 channel function in HUVECs, it had no effect on ATP release from HUVECs or U937-HUVECs adhesion. CONCLUSIONS: We demonstrated that sufentanil application increases monocyte-endothelial adherence which was associated with reduced release of ATP from Cx43 channels in monocytes. This side-effect of sufentanil should be considered seriously by clinicians.


Assuntos
Adesão Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Sufentanil/efeitos adversos , Trifosfato de Adenosina/metabolismo , Analgésicos Opioides/efeitos adversos , Conexina 43/metabolismo , Células Endoteliais/citologia , Fentanila/efeitos adversos , Células Endoteliais da Veia Umbilical Humana , Humanos , Monócitos/citologia , Remifentanil/efeitos adversos , Células U937
13.
Nutrients ; 13(5)2021 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-34064981

RESUMO

Alcoholic liver disease (ALD) is one type of liver disease, causing a global healthcare problem and mortality. The liver undergoes tissue damage by chronic alcohol consumption because it is the main site for metabolism of ethanol. Chronic alcohol exposure progresses from alcoholic fatty liver (AFL) to alcoholic steatohepatitis (ASH), which further lead to fibrosis, cirrhosis, and even hepatocellular cancer. Therapeutic interventions to combat ALD are very limited such as use of corticosteroids. However, these therapeutic drugs are not effective for long-term usage. Therefore, additional effective and safe therapies to cope with ALD are urgently needed. Previous studies confirmed that edible food plants and their bioactive compounds exert a protective effect against ALD. In this review article, we summarized the hepatoprotective potential of edible food plants and their bioactive compounds. The underlying mechanism for the prevention of ALD by edible food plants was as follows: anti-oxidation, anti-inflammation, lipid regulation, inhibition of apoptosis, gut microbiota composition modulation, and anti-fibrosis.


Assuntos
Hepatopatias Alcoólicas/terapia , Plantas Comestíveis/química , Polifenóis/uso terapêutico , Substâncias Protetoras/uso terapêutico , Consumo de Bebidas Alcoólicas , Animais , Etanol/efeitos adversos , Etanol/metabolismo , Fígado Gorduroso Alcoólico/metabolismo , Fígado Gorduroso Alcoólico/terapia , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Hepatopatias Alcoólicas/metabolismo , Neoplasias Hepáticas , Compostos Fitoquímicos/química , Compostos Fitoquímicos/uso terapêutico , Extratos Vegetais/uso terapêutico , Substâncias Protetoras/química
14.
Br J Pharmacol ; 178(18): 3783-3796, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33959955

RESUMO

BACKGROUND AND PURPOSE: Macrophages regulate iron homeostasis in the liver and play important role in hepatic ischaemia/reperfusion (I/R) injury. This study investigates the role of macrophages in iron overload-related hepatocyte damage during liver I/R. EXPERIMENTAL APPROACH: Liver biopsies from patients undergoing partial hepatectomy with or without hepatic portal occlusion were recruited and markers of hepatocyte cell death and macrophage extracellular traps (METs) were detected. A murine hepatic I/R model was also established in high-iron diet-fed mice. Ferrostatin-1 and deferoxamine were administered to investigate the role of ferroptosis in hepatic I/R injury. The macrophage inhibitor liposome-encapsulated clodronate was used to investigate the interaction between macrophages and ferroptosis. AML12 hepatocytes and RAW264.7 macrophages were co-cultured in vitro. An inhibitor of macrophage extracellular traps was used to evaluate the role and mechanism of these traps and ferroptosis in hepatic I/R injury. KEY RESULTS: Hepatocyte macrophage extracellular trap formation and ferroptosis were greater in patients who underwent hepatectomy with hepatic portal occlusion and in mice subjected to hepatic I/R. Macrophage extracellular traps increased when macrophages were subjected to hypoxia/reoxygenation and when they were co-cultured with hepatocytes. Ferroptosis increased and post-hypoxic hepatocyte survival decreased, which were reversed by inhibition of macrophage extracellular traps. Ferroptosis inhibition attenuated post-ischaemic liver damage. Moreover, iron overload induced hepatic ferroptosis and exacerbated post-ischaemic liver damage, which were reversed by the iron chelator. CONCLUSION AND IMPLICATIONS: Macrophage extracellular traps are in volved in regulating ferroptosis highlighting the therapeutic potential of macrophage extracellular traps and ferroptosis inhibition in reducing liver I/R injury.


Assuntos
Armadilhas Extracelulares , Sobrecarga de Ferro , Traumatismo por Reperfusão , Animais , Humanos , Fígado , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL
15.
Ann Transl Med ; 9(3): 234, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33708861

RESUMO

BACKGROUND: Gestational diabetes mellitus (GDM) is among the most common metabolic diseases during pregnancy and inevitably leads to maternal and fetal complications. Hyperglycemia results in injury to vascular endothelial cells, including monocyte-endothelial adhesion, which is considered to be the initiating factor of vascular endothelial cell injury. Connexin 43 (Cx43) plays a key role in this adhesion process. Therefore, this study aimed to explore the effects of Cx43 on monocyte-endothelial adhesion in GDM-induced injury of vascular endothelial cells. METHODS: Human umbilical vein endothelial cells (HUVECs) were isolated from umbilical cords from pregnant women with and without GDM. THP-1 cells (a human leukemia monocytic cell line) adhering to HUVECs, related molecules [intracellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1)], and the activity of the phosphoinositide 3-kinase/protein kinase B/Nuclear factor- kappa B (PI3K/AKT/NF-κB) signaling pathway were compared between the normal and GDM-HUVECs. Oleamide and specific small interfering ribonucleic acids (siRNAs) were used to inhibit Cx43 expression in GDM-HUVECs to observe the effects of Cx43 on the adhesion of THP-1 cells and HUVECs. RESULTS: A much higher number of THP-1 cells adhered to GDM-HUVECs than to normal HUVECs. This was accompanied by an increased expression of Cx43, ICAM-1, and VCAM-1, as well as activation of the PI3K/AKT/NF-κB signaling pathway. After the inhibition of Cx43 expression in GDM-HUVECs with oleamide and specific siRNA, THP-1-HUVEC adhesion, ICAM-1 and VCAM-1 expression, and activation of PI3K/AKT/NF-κB signaling pathway were all attenuated. Hyperglycemia was able to increase expression of Cx43 in HUVECs. CONCLUSIONS: For the first time, Cx43 expression was found to be substantially higher in GDM-HUVECs than in normal HUVECs. Hyperglycemia caused the overexpression of Cx43 in HUVECs, which resulted in the activation of the PI3K/AKT/NF-κB signaling pathway and the increase of its downstream adhesion molecules, including ICAM-1 and VCAM-1, ultimately leading to increased monocyte-endothelial adhesion.

16.
Materials (Basel) ; 14(3)2021 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-33498490

RESUMO

Durability is one important problem that pavement engineers need to address in pavement's long service life. Furthermore, easily recycled pavement materials, and safe and efficient pavement construction are also important areas for development in road engineering. For these reasons, a new asphalt steel plastic (ASP) pavement structure was proposed with an asphalt mixture forming the surface layer, and steel plate and plastic materials functioning as the main load-bearing layers. Based on a comprehensive performance review and cost-benefit analysis, stone mastic asphalt (SMA) is recommended to be used as the surface layer; and A656 steel plate and acrylonitrile butadiene styrene (ABS) plastic materials should be the main load-bearing layer, on top of a foundation layer made with graded crushed stones. A glass fiber reinforced polymer (GFRP) insulation layer is recommended for use between the steel plate and ABS. Mechanical properties of the ASP pavement were analyzed using the finite element method. Laboratory tests were conducted to verify the thermal insulation performance of GFRP, the high-temperature stability and the fatigue resistance of ASP pavement. Results show that some of the mechanical properties of ASP pavement (with a structure of 80 mm SMA asphalt mixture, 8 mm steel plate, 140 mm ABS and 200 mm crushed stones) are comparable with conventional long-life pavement (with 350 mm asphalt layer overlaying 400 mm graded crushed stones). Dynamic stability of the ASP slab specimens can reach 10,000 times/mm, and the fatigue life is about twice that of SMA. Besides, the ASP pavement can be prefabricated and assembled on-site, and thus can greatly improve construction efficiency. From the lifecycle perspective, ASP pavement has many advantages over traditional pavements, such as durability, lower environmental footprint and recyclability, making it is worth further research.

17.
Life Sci ; 272: 119119, 2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-33508296

RESUMO

AIMS: Acute kidney injury (AKI) is a severe complication of autologous orthotopic liver transplantation (AOLT). Apoptosis has been shown to be involved in renal ischemia/reperfusion, and the PI3K/AKT signaling pathway is involved in numerous cell processes, including promoting cell survival and inhibiting apoptosis. We aimed to verify whether the PI3K/AKT signaling pathway participates in the development of post-AOLT AKI. METHODS: Male Sprague-Dawley rats underwent AOLT with or without treatment with insulin-like growth factor-1 (IGF-1, a PI3K/AKT activator) and LY294002 (a PI3K/AKT inhibitor; n = 8/group). NRK-52E cells (rat renal tubular epithelial cell line) were subjected to hypoxia-re-oxygenation to mimic renal cell I/R injury in vitro, and confirm whether silencing information regulator 1 (SIRT1) mediated the protective effects of PI3K/AKT by deacetylating forkhead protein O3a (FoxO3a). KEY FINDINGS: During the reperfusion stage, kidney injury peaked at 8 h after reperfusion, then gradually recovered, which was consistent with the dynamic changes in apoptosis and the protein expressions of Bcl-2 interacting mediator of cell death (Bim), Fas ligand (FasL), and nuclear FoxO3a AKT phosphorylation and nuclear SIRT1 protein expression were also upregulated. IGF-1 application decreased Bim, FasL, and nuclear FoxO3a protein expressions, and protected against apoptosis and AKI. In NRK-52E cells, IGF-1 upregulated nuclear SIRT1 expression, reduced FoxO3a acetylation, downregulated Bim and FasL protein expressions, and attenuated apoptosis and AKI; these effects were reversed by SIRT1 blocking. CONCLUSION: The activation of the PI3K/AKT signaling pathway not only induced FoxO3a nuclear export but also deacetylation through upregulating nuclear SIRT1 expression to attenuate post-AOLT AKI.


Assuntos
Injúria Renal Aguda/metabolismo , Proteína Forkhead Box O3/metabolismo , Acetilação , Transporte Ativo do Núcleo Celular , Injúria Renal Aguda/fisiopatologia , Animais , Apoptose/efeitos dos fármacos , China , Proteína Forkhead Box O3/fisiologia , Transplante de Fígado/efeitos adversos , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
18.
Biochem Biophys Res Commun ; 530(1): 54-59, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32828315

RESUMO

In this study, the regulation of miR-15b-5p on myocardial ischemia reperfusion (I/R) injury-induced arrhythmia and myocardial apoptosis was investigated in mice. We observed the change in miR-15b-5p expression after mice suffered from myocardial I/R injury and the change in myocardial injury, infarct size, apoptosis, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), superoxide dismutase (SOD) and malondialdehyde (MDA) after down-regulation of miR-15b-5p expression. The negative regulation of miR-15b-5p to KCNJ2 as well as whether cardioprotective effect formed by miR-15b-5p down-regulation relied on the increase of KNCJ2 expression were measured by dual-luciferase reporter assay system. miR-15b-5p expression increased and KCNJ2 mRNA and protein expressions decreased after myocardial ischemia reperfusion (all P < 0.05). miR-15b-5p negatively regulated KCNJ2 in a targeted way. Down-regulating miR-15b-5p expression or increasing KCNJ2 expression significantly decreased the incidence of arrhythmia, infarct size and apoptosis after myocardial I/R and lowered MDA content in the myocardial tissue as well as IL-6 and TNF-α content in the blood (all P < 0.05). KCNJ2 gene knockout reversed the above cardioprotective effect formed by miR-15b-5p down-regulation (P < 0.05). Down-regulating miR-15b-5p expression or up-regulating KCNJ2 expression improves arrhythmia after mice suffered from myocardial I/R injury and inhibits myocardial apoptosis.


Assuntos
Apoptose , Arritmias Cardíacas/genética , Regulação para Baixo , MicroRNAs/genética , Traumatismo por Reperfusão Miocárdica/genética , Animais , Arritmias Cardíacas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/patologia
19.
Med Sci Monit ; 26: e923758, 2020 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-32541641

RESUMO

BACKGROUND The purpose of this study was to investigate factors influencing bleeding in patients with acute coronary syndrome (ACS) who are on aspirin and ticagrelor as dual antiplatelet therapy. MATERIAL AND METHODS This retrospective case-control study included 50 patients with ACS (25 with reported bleeding events and 25 without) on aspirin and ticagrelor. Adenosine diphosphate (ADP)- and arachidonic acid (ACA)-induced platelet aggregation rates were measured using light transmission aggregometry. Single-nucleotide polymorphisms (SNPs) in PEAR1, GP1BA, and GSTP1 were genotyped. RESULTS ACA-induced platelet aggregation rates were obviously lower in patients with bleeding events than in those without (13.28±8.46% vs. 24.93±9.89%, P<0.001). No significant differences in ADP-induced platelet aggregation rates were observed between the 2 groups (16.17±9.74% vs. 16.88±12.69%, P>0.05). Among those with bleeding events and among controls, 70% and 80% had an ACA-induced platelet aggregation rate of 0-18% and 18-50%, respectively. Mutation rates of rs6065 in GP1BA and rs1695, rs4891, and rs8191439 in GSTP1 also differed significantly between the 2 groups. CONCLUSIONS Lower ACA-induced platelet aggregation rates are associated with increased risk of bleeding in patients with ACS who are on aspirin and ticagrelor. An ACA-induced platelet aggregation rate of 18% may be considered the cutoff point for identifying high risk of aspirin-associated bleeding events in patients with ACS. SNP genotyping may also help predict the risk of bleeding in patients with ACS.


Assuntos
Síndrome Coronariana Aguda/terapia , Aspirina/efeitos adversos , Hemorragia/genética , Intervenção Coronária Percutânea , Inibidores da Agregação Plaquetária/efeitos adversos , Agregação Plaquetária , Ticagrelor/efeitos adversos , Difosfato de Adenosina , Idoso , Ácido Araquidônico , Estudos de Casos e Controles , Terapia Antiplaquetária Dupla/efeitos adversos , Feminino , Genótipo , Glutationa S-Transferase pi/genética , Hemorragia/sangue , Hemorragia/induzido quimicamente , Humanos , Masculino , Pessoa de Meia-Idade , Complexo Glicoproteico GPIb-IX de Plaquetas/genética , Polimorfismo de Nucleotídeo Único , Receptores de Superfície Celular/genética , Estudos Retrospectivos
20.
Eur J Pharmacol ; 876: 173056, 2020 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-32147436

RESUMO

Hepatic ischemia/reperfusion (I/R) injury is a common complication in the clinical setting. Our previous study has shown that connexin 32 (Cx32) plays a major role in renal I/R injury; however, the role of Cx32 in hepatic I/R injury remains unknown. Liver tissue and serum samples from patients undergoing orthotopic liver transplantation (OLT) were used to evaluate the function of Cx32 in OLT post-reperfusion injury. Then, partial hepatic ischemia was established in global Cx32 knockout mice and wild-type mice followed by reperfusion. Hepatic injury markers were examined. Cx32 small interfering RNA and the p53 inhibitor, pifithrin-α, tenovin-1 were used to examine the relationship between Cx32 and the p53/puma pathways in the BRL-3A and murine primary hepatocytes hypoxia/reoxygenation (H/R) model. Corresponding to liver damage, Cx32 was significantly induced both during OLT in human patients and partial hepatic I/R in mice. Cx32 KO mice exhibited less liver injury than controls. Cx32 deficiency significantly suppressed the p53/puma pathways and hepatocyte apoptosis. Similar results were observed in the BRL-3A and murine primary hepatocytes H/R model. Propofol protected against OLT post-reperfusion injury and hepatocyte apoptosis by inhibiting Cx32. In conclusion Cx32 is a novel regulator of hepatic I/R injury through the modulation of hepatocyte apoptosis and damage, largely via the p53/puma signaling pathway.


Assuntos
Conexinas/antagonistas & inibidores , Isquemia/prevenção & controle , Transplante de Fígado , Fígado/irrigação sanguínea , Propofol/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Conexinas/deficiência , Conexinas/genética , Modelos Animais de Doenças , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Isquemia/metabolismo , Isquemia/patologia , Pós-Condicionamento Isquêmico , Fígado/metabolismo , Fígado/patologia , Testes de Função Hepática , Camundongos Endogâmicos C57BL , Camundongos Knockout , Propofol/administração & dosagem , Estudos Prospectivos , Ratos , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteína beta-1 de Junções Comunicantes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA