Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Front Pharmacol ; 13: 967412, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36105229

RESUMO

Aims: To explore the population pharmacokinetics of colistin sulfate and to optimize the dosing strategy for critically ill patients. Methods: The study enrolled critically ill adult patients who received colistin sulfate intravenously for more than 72 h with at least one measurement of plasma concentration. Colistin concentrations in plasma or urine samples were measured by ultraperformance liquid chromatography tandem mass spectrometry (LC-MS/MS). The population pharmacokinetics (PPK) model for colistin sulfate was developed using the Phoenix NLME program. Monte Carlo simulation was conducted to evaluate the probability of target attainment (PTA) for optimizing dosing regimens. Results: A total of 98 plasma concentrations from 20 patients were recorded for PPK modeling. The data were adequately described by a two-compartment model with linear elimination. During modeling, creatinine clearance (CrCL) and alanine aminotransferase (ALT) were identified as covariates of the clearance (CL) and volume of peripheral compartment distribution (V2), respectively. In addition, colistin sulfate was predominantly cleared by the nonrenal pathway with a median urinary recovery of 10.05% with large inter-individual variability. Monte Carlo simulations revealed a greater creatinine clearance associated with a higher risk of sub-therapeutic exposure to colistin sulfate. The target PTA (≥90%) of dosage regimens recommended by the label sheet was achievable only in patients infected by pathogens with MIC ≤0.5 mg/L or with renal impairments. Conclusion: Our study showed that the dose of intravenous colistin sulfate was best adjusted by CrCL and ALT. Importantly, the recommended dosing regimen of 1.0-1.5 million units daily was insufficient for patients with normal renal functions (CrCL ≥80 ml/min) or those infected by pathogens with MIC ≥1.0 mg/L. The dosage of colistin sulfate should be adjusted according to renal function and drug exposure.

2.
Inflamm Res ; 71(9): 1067-1078, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35900381

RESUMO

BACKGROUND: Acute pancreatitis (AP) is an inflammatory process of the pancreas resulting from biliary obstruction or alcohol consumption. Approximately, 10-20% of AP can evolve into severe AP (SAP). In this study, we sought to explore the physiological roles of the transcription factor serum response factor (SRF), annexin A2 (ANXA2), and nuclear factor-kappaB (NF-κB) in SAP. METHODS: C57BL/6 mice and rat pancreatic acinar cells (AR42J) were used to establish an AP model in vivo and in vitro by cerulein with or without lipopolysaccharide (LPS). Production of pro-inflammatory cytokines (IL-1ß and TNF-α) were examined by ELISA and immunoblotting analysis. Hematoxylin and eosin (HE) staining and TUNEL staining were performed to evaluate pathological changes in the course of AP. Apoptosis was examined by flow cytometric and immunoblotting analysis. Molecular interactions were tested by dual luciferase reporter, ChIP, and Co-IP assays. RESULTS: ANXA2 was overexpressed in AP and correlated to the severity of AP. ANXA2 knockdown rescued pancreatic acinar cells against inflammation and apoptosis induced by cerulein with or without LPS. Mechanistic investigations revealed that SRF bound with the ANXA2 promoter region and repressed its expression. ANXA2 could activate the NF-κB signaling pathway by inducing the nuclear translocation of p50. SRF-mediated transcriptional repression of ANXA2-protected pancreatic acinar cells against AP-like injury through repressing the NF-κB signaling pathway. CONCLUSION: Our study highlighted a regulatory network consisting of SRF, ANXA2, and NF-κB that was involved in AP progression, possibly providing some novel targets for treating SAP.


Assuntos
Anexina A2/metabolismo , Pancreatite , Fator de Resposta Sérica/metabolismo , Doença Aguda , Animais , Anexina A2/genética , Ceruletídeo/efeitos adversos , Ceruletídeo/metabolismo , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Pâncreas/patologia , Pancreatite/induzido quimicamente , Pancreatite/patologia , Ratos , Transdução de Sinais
3.
Kidney Dis (Basel) ; 8(2): 126-136, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35527991

RESUMO

Background: Hydrogen is a chemical substance that has yet to be widely used in medicine. However, recent evidence indicates that hydrogen has multi-faceted pharmacological effects such as antioxidant, anti-inflammatory, and antiapoptotic properties. An increased number of studies are being conducted on the application of hydrogen in various diseases, especially those affecting the renal system. Summary: Hydrogen can be inhaled, as a gas or liquid, and can be administered orally, intravenously, or locally. Hydrogen can rapidly enter suborganelles such as mitochondria and nucleus by simple diffusion, producing reactive oxygen species (ROS) and triggering DNA damage. Hydrogen can selectively scavenge hydroxyl radical (•OH) and peroxynitrite (ONOO-), but not other reactive oxygen radicals with physiological functions, such as peroxyanion (O2-) and hydrogen peroxide (H2O2). Although the regulatory effect of hydrogen on the signal transduction pathway has been confirmed, the specific mechanism of its influence on signal molecules remains unknown. Although many studies have investigated the therapeutic and preventive effects of H2 in cellular and animal experiments, clinical trials are few and still far behind. As a result, more clinical trials are required to investigate the role of hydrogen in kidney disease, as well as the effect of its dose, timing, and form on the overall efficacy. Large-scale randomized controlled clinical trials will be required before hydrogen can be used to treat renal illnesses. Key Messages: This article reviews the mechanisms of hydrogen in the treatment of renal disease and explores the possibilities of its use in clinical practice.

4.
Mol Ther Nucleic Acids ; 25: 502-514, 2021 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-34589273

RESUMO

Naringin exhibited various pharmacological activities. However, its biological function and underlying mechanism in regulating macrophage polarization remain elusive. This study aimed to investigate the regulatory network between naringin and macrophage polarization in sepsis-induced intestinal injury. Cecal ligation and puncture (CLP) was used to establish the animal model of sepsis. Chromatin immunoprecipitation and a luciferase reporter assay were used to determine the interplay between peroxisome proliferator-activated receptor γ (PPARγ) and miR-21 promoter, as well as miR-21 and its target genes. Naringin enhanced the overall survival of septic mice and alleviated the CLP-induced inflammatory response and intestinal damage. This was accompanied by the increased expression of PPARγ in the intestines and the stimulation of ileal macrophages toward the M2 phenotype. Furthermore, in lipopolysaccharide-stimulated bone marrow-derived macrophages, naringin stimulated M2 polarization. Mechanistically, PPARγ inhibition attenuated the promotion of M2 polarization caused by naringin, and the naringin/PPARγ regulatory work was compromised by miR-21 inhibition. The present study suggested that naringin promoted M2 polarization via the PPARγ/miR-21 axis, thus relieving sepsis-induced intestinal injury. This study provides novel insights into the mechanism by which naringin alleviated sepsis-induced intestinal injury through regulation of macrophage polarization.

5.
Autoimmunity ; 54(7): 460-470, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34402705

RESUMO

BACKGROUND: Severe acute pancreatitis (SAP) is a common acute abdominal disease with high morbidity and mortality. However, the mechanism underlying SAP is still unclear. METHODS: Cerulean and LPS (Cer-LPS) was used to establish a rat model and an in vitro model of SAP. qRT-PCR, western blot and IHC were determined to analyse the expression of mRNA and proteins. IL-1ß, TNF-α and IL-6 levels were measured applying ELISA. H&E staining was determined to observe the pathological changes. Apoptosis was tested by AV-PI staining using flow cytometry. CCK8 assay was taken to detect cell viability. Cell migration was assessed by transwell assay. Tube formation assay was conducted to evaluate angiogenesis. Luciferase assay was used to detect relationship of miR-20b-5p and AKT3. RESULTS: MiR-20b-5p was lowly expressed in SAP models both in vivo and in vitro. Overexpression of miR-20b-5p restrained inflammation and apoptosis in Cer-LPS treated pancreatic acinar cells. Furthermore, miR-20b-5p promoted the angiogenesis of vascular endothelial cells, since the viability, migration and the capability of tube formation were increased by miR-20b-5p. Mechanically, miR-20b-5p directly targeted AKT3 to promote autophagy. Furthermore, miR-20b-5p could prevent the inflammation, apoptosis and enhance angiogenesis via enhancing autophagy, which was verified in vivo. CONCLUSION: This study demonstrated miR-20b-5p attenuates SAP through directly targeting AKT3 to regulate autophagy, subsequently inhibit inflammation and apoptosis, and promote angiogenesis. Our findings suggested a novel target of miR-20b-5p for the therapy of SAP.


Assuntos
MicroRNAs , Pancreatite , Doença Aguda , Animais , Apoptose/genética , Autofagia/genética , Células Endoteliais/metabolismo , Inflamação/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Neovascularização Fisiológica , Pancreatite/genética , Proteínas Proto-Oncogênicas c-akt , Ratos
6.
Inflammation ; 44(3): 999-1013, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33417179

RESUMO

Acute pancreatitis (AP) is an inflammatory, complicated pancreatic disease, carrying significant morbidity and mortality. However, the molecular and cellular mechanisms involved in AP pathogenesis remain to be elucidated. Here, we explore the role of FOXF1 adjacent non-coding developmental regulatory RNA (FENDRR) in AP progression. Caerulein with or without LPS- induced or taurolithocholic acid 3-sulfate (TLC-S)-induced AP mouse models and cell models were performed for the validation of FENDRR expression in vivo and in vitro, respectively. Histopathological examinations of pancreatic tissues were performed to evaluate the severity of AP. Transmission electron microscopy was utilized to visualize the autophagic vacuoles. siRNA specifically targeting FENDRR was further applied. Flow cytometry was employed to assess cell apoptosis. ELISA, immunoflureoscence, and western blotting analysis were also performed to determine the levels of inflammatory cytokines and autophagy activity. RNA immunoprecipitation (RIP) and chromatin immunoprecipitation (ChIP) assays were carried out to reveal the epigenetic regulation of FENDRR on ATG7. Additionally, silencing FENDRR was also verified in AP mouse models. Higher FENDRR and impaired autophagy were displayed in both AP mouse models and cell models. FENDRR knockdown dramatically attenuated caerulein- or TLC-S-induced AR42J cells apoptosis and autophagy suppression. Further mechanistic experiments implied that the action of FENDRR is moderately attributable to its repression of ATG7 via direct interaction with the epigenetic repressor PRC2. Moreover, the silencing of FENDRR significantly induced the promotion of ATG7, thus alleviating the development of AP in vivo. Our study highlights FENDRR as a novel target that may contribute to AP progression, suggesting a therapeutic target for AP treatment.


Assuntos
Proteína 7 Relacionada à Autofagia/metabolismo , Autofagia , Epigênese Genética , Pâncreas/metabolismo , Pancreatite/metabolismo , Complexo Repressor Polycomb 2/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Proteína 7 Relacionada à Autofagia/genética , Linhagem Celular , Ceruletídeo , Citocinas/metabolismo , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos , Masculino , Camundongos Endogâmicos C57BL , Pâncreas/ultraestrutura , Pancreatite/induzido quimicamente , Pancreatite/genética , Pancreatite/patologia , Complexo Repressor Polycomb 2/genética , RNA Longo não Codificante/genética , Transdução de Sinais
7.
Pharm Biol ; 58(1): 898-904, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32893702

RESUMO

CONTEXT: Sepsis is currently one of the leading causes of death in intensive care units (ICUs). Sesamin was previously reported to inhibit inflammation. However, no studies have revealed the impact of sesamin on sepsis. OBJECTIVE: We studied the mechanism underlying the effect of sesamin on the pathophysiology of sepsis through the HMGB1/TLR4/IL-33 signalling pathway. MATERIALS AND METHODS: Fifty male BALB/c mice (n = 10 per group) were used to establish a caecal ligation and puncture (CLP) mouse model, and given daily injections of sesamin at a low, middle, or high concentration (25, 50, or 100 µM) during the seven-day study period; survival curves were generated by the Kaplan-Meier method. H&E staining and TUNEL staining were performed to assess changes in intestinal morphology intestinal damage in the mouse intestinal epithelium. Molecules related to the HMGB1/TLR4/IL-33 pathway were assessed by RT-qPCR and Western blotting. RESULTS: We found mice in the sepsis group survived for only 4 days, while those treated with sesamin survived for 6-7 days. In addition, sesamin significantly relieved the increase in the levels of MPO (21%, 33.3%), MDA (40.5% and 31.6%), DAO (1.24-fold and 2.31-fold), and pro-inflammatory cytokines such as TNF-α (75% and 79%) and IL-6 (1-fold and 1.67-fold) 24 and 48 h after sepsis induction and downregulated the expression of HMGB1, TLR4, and IL-33 while upregulating the expression of ZO-1 and occludin. DISCUSSION AND CONCLUSIONS: Sesamin improved the 7-day survival rate of septic mice, suppressed the inflammatory response in sepsis through the HMGB-1/TLR4/IL-33 signalling pathway, and further alleviated intestinal injury.


Assuntos
Dioxóis/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/lesões , Lignanas/farmacologia , Sepse/tratamento farmacológico , Animais , Bactérias/efeitos dos fármacos , Linhagem Celular , Citocinas/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Células Epiteliais , Proteína HMGB1/metabolismo , Humanos , Inflamação/tratamento farmacológico , Interleucina-33/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ocludina/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo
8.
Pancreas ; 49(4): 543-551, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32282768

RESUMO

OBJECTIVES: MicroRNAs have been considered to be closely related with the development of severe acute pancreatitis (SAP), and microRNA-375 (miR-375) was believed to be a marker of SAP. We aim to investigate the role of miR-375 in regulating SP. METHODS: Cerulein and lipopolysaccharide were used to establish the models of SAP. AR42J cell line was chosen for study in vitro. Flow cytometry was applied for assessing apoptosis. The contents of inflammatory factors were detected with related enzyme-linked immunosorbent assay and quantitative real-time polymerase chain reaction assays. Hematoxylin and eosin staining was applied to observe the pathological changes of pancreatic tissues. Immunohistochemistry analysis was conducted for investigating the expression of light chain 3. RESULTS: The level of miR-375 in pancreatitis tissues and cell lines was upregulated. Overexpression of miR-375 promoted inflammation and the apoptosis of acinar cells through inhibiting autophagy. The binding site between miR-375 and ATG7 was identified, and miR-375 could directly regulate the ATG7. microRNA-375 suppressed autophagy and promoted inflammation and the apoptosis of acinar cells via targeting ATG7. CONCLUSIONS: We proved that miR-375 could inhibit autophagy and promote inflammation and the apoptosis of acinar cells through regulating ATG7. This study first proves that miR-375 modulates the development of SAP through targeting ATG7.


Assuntos
Células Acinares/patologia , Proteína 7 Relacionada à Autofagia/antagonistas & inibidores , Autofagia/genética , MicroRNAs/genética , Pancreatite/genética , Células Acinares/metabolismo , Animais , Apoptose/genética , Proteína 7 Relacionada à Autofagia/genética , Sítios de Ligação , Linhagem Celular , Ceruletídeo/toxicidade , Modelos Animais de Doenças , Humanos , Lipopolissacarídeos/toxicidade , MicroRNAs/biossíntese , Pancreatite/induzido quimicamente , Pancreatite/metabolismo , Pancreatite/patologia , Ligação Proteica , Estabilidade de RNA/genética , RNA Mensageiro/metabolismo , Ratos , Regulação para Cima
9.
IUBMB Life ; 72(3): 476-491, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31846199

RESUMO

OBJECTIVE: Many studies have reported the correlation of microRNAs (miRNAs) with cancers, yet few have proposed the function of miR-150 in gastric cancer. This study intends to discuss the role of miR-150 in gastric cancer development by regulating IRX1. METHODS: Gastric cancer tissues and adjacent tissues were collected. MiR-150-3p, IRX1, CXCL14, and NF-κB (p65) expressions were detected. Gastric cancer cell lines SNU-1 and MKN-45 were used for subsequent cellular experiments. Cell proliferation, colony formation, migration and invasion, apoptosis, and cell cycle distribution in SNU-1 and MKN-45 cells were determined via gain-of and loss-of-function assays. The tumor growth in nude mice was also detected. RESULTS: MiR-150, CXCL14, and NF-κB (p65) were upregulated and IRX1 was downregulated in gastric cancer tissues and cells. CXCL14 and NF-κB (p65) expression was positively related to miR-150 expression and negatively to IRX1 expression. MiR-150 inhibition and IRX1 overexpression in SNU-1 cells restricted viability, colony formation, migration, and invasion abilities, but boosted apoptosis of gastric cancer cells in vitro, and also repressed tumor growth in vivo. These results could be reversed by miR-150 elevation and IRX1 silencing, and the results from in vivo and in vitro experiments were consistent. CONCLUSION: Our study reveals that miR-150 downregulation restrains proliferation, migration, and invasion, while facilitating apoptosis of gastric cancer cells by upregulating IRX1.


Assuntos
Proteínas de Homeodomínio/genética , MicroRNAs/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Fatores de Transcrição/genética , Adulto , Idoso , Animais , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Quimiocinas CXC/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos Endogâmicos BALB C , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Fator de Transcrição RelA/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Life Sci ; 228: 11-20, 2019 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-30690081

RESUMO

BACKGROUND: Sepsis is a life-threatening organ dysfunction disease caused by a dysregulated host response to infection. Rutaecarpine is an important alkaloid component of Evodia rutaecarpa. There has been no study on the therapeutic effects of rutaecarpine in sepsis. METHODS: Mice were randomly assigned into four groups: sham, sepsis, sepsis plus vehicle and sepsis plus rutaecarpine groups. Mice in sepsis were administered CLP surgery. Rutaecarpine or vehicle was injected intraperitoneally 1 h after CLP. The liver damage, bacterial infection, survival rate and weight loss were observed, and changes in the ratio of peritoneal resident macrophages were analyzed by flow cytometry and immunofluorescence microscopy. Western blotting was used to identify the levels of NF-κB signaling pathway, ER stress and apoptosis related proteins. TUNEL and Annexin V/PI assay were used to detect the apoptosis of liver tissues and peritoneal resident macrophages, respectively. ELISA and qRT-PCR were used to detect the inflammatory factors. RESULTS: Rutaecarpine alleviated weight loss, bacterial infection and liver injury, and regulated inflammation homeostasis, enhancing survival rate induced by sepsis. Population of peritoneal resident macrophages (CD11b+F4/80hiMHCIIlow) was significantly decreased in sepsis mice, which was resulted from ER stress-induced apoptosis through caspase-12 signaling pathway. Rutaecarpine restored the ratio of peritoneal resident macrophages and the level of GATA6 in CD11b+ peritoneal macrophages. Rutaecarpine could also attenuate sepsis-induced inflammatory responses through inhibiting the activation of ER stress/NF-κB pathway. CONCLUSION: Rutaecarpine ameliorated sepsis-induced peritoneal resident macrophages apoptosis and inflammation responses through inhibition of ER stress-mediated caspase-12 and NF-κB pathways. Our study provided new insights for drug development against sepsis.


Assuntos
Anti-Inflamatórios/uso terapêutico , Apoptose/efeitos dos fármacos , Alcaloides Indólicos/uso terapêutico , Inflamação/tratamento farmacológico , Macrófagos Peritoneais/efeitos dos fármacos , Quinazolinas/uso terapêutico , Sepse/tratamento farmacológico , Animais , Anti-Inflamatórios/química , Evodia/química , Alcaloides Indólicos/química , Inflamação/complicações , Inflamação/imunologia , Macrófagos Peritoneais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/imunologia , Quinazolinas/química , Sepse/complicações , Sepse/imunologia , Transdução de Sinais/efeitos dos fármacos
11.
Biomed Pharmacother ; 103: 50-58, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29635128

RESUMO

BACKGROUND: Sepsis is commonly associated with excessive stimulation of host immune system and result in multi-organ failure dysfunction. Naringin has been reported to exhibit a variety of biological effects. The present study aimed to investigate the protective effect of naringin on sepsis-induced injury of intestinal barrier function in vivo and in vitro. METHODS: Mice were randomly divided into 4 groups named sham (n = 20), CLP + vehicle (n = 20), CLP + NG (30 mg/kg) (n = 20) and CLP + NG (60 mg/kg) (n = 20) groups. Sepsis was induced by cecal ligation and puncture (CLP). H&E staining and transmission electron microscopy (TEM) were performed to observe intestinal mucosal morphology. ELISA was used to determine the intestinal permeability and inflammatory response in vivo and in vitro. Western blot and RhoA activity assay were performed to determine the levels of tight junction proteins and the activation of indicated signaling pathways. MTT assay was used to determine cell viability. RESULTS: Naringin improved survival rate of CLP mice and alleviated sepsis-induced intestinal mucosal injury. Furthermore, naringin improved impaired intestinal permeability and inhibited the release of TNF-α and IL-6, while increased IL-10 level in CLP mice and lipopolysaccharide (LPS)-stimulated MODE-K cells in a dose-dependent manner. Naringin increased the expression of tight junction proteins ZO-1 and claudin-1 via RhoA/ROCK/NF-κB/MLCK/MLC signaling pathway in vivo and in vitro. CONCLUSION: Naringin improved sepsis-induced intestinal injury via RhoA/ROCK/NF-κB/MLCK/MLC signaling pathway in vivo and in vitro.


Assuntos
Flavanonas/farmacologia , Intestinos/lesões , Intestinos/patologia , Cadeias Leves de Miosina/metabolismo , NF-kappa B/metabolismo , Sepse/patologia , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Ceco/patologia , Sobrevivência Celular/efeitos dos fármacos , Claudina-1/metabolismo , Flavanonas/administração & dosagem , Inflamação/patologia , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Ligadura , Lipopolissacarídeos , Masculino , Camundongos , Quinase de Cadeia Leve de Miosina/metabolismo , Permeabilidade , Fosforilação/efeitos dos fármacos , Punções , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida , Fator de Necrose Tumoral alfa/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo
12.
Angew Chem Int Ed Engl ; 56(52): 16606-16610, 2017 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-29135065

RESUMO

Lithium-ion batteries (LIBs) are being used to power the commercial electric vehicles (EVs). However, the charge/discharge rate and life of current LIBs still cannot satisfy the further development of EVs. Furthermore, the poor low-temperature performance of LIBs limits their application in cold climates and high altitude areas. Herein, a simple prelithiation method is developed to fabricate a new LIB. In this strategy, a Li3 V2 (PO4 )3 cathode and a pristine hard carbon anode are used to form a primary cell, and the initial Li+ extraction from Li3 V2 (PO4 )3 is used to prelithiate the hard carbon. Then, the self-formed Li2 V2 (PO4 )3 cathode and prelithiated hard carbon anode are used to form a 4 V LIB. The LIB exhibits a maximum energy density of 208.3 Wh kg-1 , a maximum power density of 8291 W kg-1 and a long life of 2000 cycles. When operated at -40 °C, the LIB can keep 67 % capacity of room temperature, which is much better than conventional LIBs.

13.
Angew Chem Int Ed Engl ; 56(31): 9126-9130, 2017 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-28612470

RESUMO

The utilization of CO2 in Li-CO2 batteries is attracting extensive attention. However, the poor rechargeability and low applied current density have remained the Achilles' heel of this energy device. The gel polymer electrolyte (GPE), which is composed of a polymer matrix filled with tetraglyme-based liquid electrolyte, was used to fabricate a rechargeable Li-CO2 battery with a carbon nanotube-based gas electrode. The discharge product of Li2 CO3 formed in the GPE-based Li-CO2 battery exhibits a particle-shaped morphology with poor crystallinity, which is different from the contiguous polymer-like and crystalline discharge product in conventional Li-CO2 battery using a liquid electrolyte. Accordingly, the GPE-based battery shows much improved electrochemical performance. The achieved cycle life (60 cycles) and rate capability (maximum applied current density of 500 mA g-1 ) are much higher than most of previous reports, which points a new way to develop high-performance Li-CO2 batteries.

14.
J Surg Res ; 211: 70-78, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28501133

RESUMO

BACKGROUND: Intravenous administration of ulinastatin (UTI), a broad spectral protease inhibitor, has been used on an experimental basis with severe sepsis patients in Asia. However, the effects of intraintestinal administration of UTI on intestinal and multiple organ damage in sepsis have not been reported. MATERIALS AND METHODS: In this study, we established a sepsis model in rats using cecal ligation and puncture and compared the effects of intraintestinal administration of UTI through an artificial fistula of duodenum and intraperitoneal administration of UTI on the pathophysiological changes of sepsis. RESULTS: It was found that intraintestinal administration of UTI (1) significantly improved the survival of septic rats, (2) significantly reduced the serum levels of tumor necrosis factor-α, interleukin-1ß, interleukin-6 as well as intestinal injury biomarkers diamine oxidase, D-lactic acid, and fluorescein isothiocyanate-dextran 4, and (3) significantly reduced intestinal microscopic and ultrastructural damage of septic rats. In addition, the protective effects of intraintestinal administration of UTI were significantly better than those of intraperitoneal administration of UTI. CONCLUSIONS: Overall, the present study for the first time revealed that intraintestinal administration of protease inhibitor UTI could reduce systemic inflammatory responses and multiple organ dysfunction in rats with sepsis by inhibiting autodigestion of intestinal wall due to proteases and provided new research ideas and experimental evidences for treatment of sepsis by intraintestinal administration of UTI.


Assuntos
Glicoproteínas/administração & dosagem , Intestinos/efeitos dos fármacos , Sepse/tratamento farmacológico , Inibidores da Tripsina/administração & dosagem , Animais , Biomarcadores/sangue , Glicoproteínas/farmacologia , Glicoproteínas/uso terapêutico , Injeções Intraperitoneais , Intestinos/patologia , Masculino , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Sepse/sangue , Sepse/mortalidade , Sepse/fisiopatologia , Resultado do Tratamento , Inibidores da Tripsina/farmacologia , Inibidores da Tripsina/uso terapêutico
15.
Shock ; 44(4): 357-64, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26125086

RESUMO

Endostatin is an endogenous inhibitor of vascular endothelium. It can inhibit endothelial cell migration, proliferation, and vascular angiogenesis and is mainly used for anticancer therapy. We have previously found that endostatin is an important node protein in the pathogenesis of sepsis. However, its impacts on sepsis have not yet been reported. We established a septic mouse model using cecal ligation and puncture (CLP) and gave the mice either endostatin or placebo (saline). The effects of endostatin on serum enzyme, Evans blue leakage, lung wet-to-dry weight ratio, and cytokine (tumor necrosis factor α, interleukin 1ß [IL-1ß], and IL-6) production were assessed. Survival rates were observed for up to 3 days. In addition, we examined the effects of endostatin on serum vascular endothelial growth factor A (VEGF-A), VEGF-C, and pathological changes and scores of lung tissues as well as the phosphorylation of JNK, p38, and ERKl/2 proteins in lung tissues of mice with sepsis. We found that endostatin can increase the survival of septic mice in a time- and dose-dependent manner probably by reducing multiorgan dysfunctions shown by serum indicators, morphologic changes, Evans blue leakage, wet-to-dry weight ratio, and inflammation of lung tissues. In addition, endostatin could reduce serum tumor necrosis factor α, IL-1ß, IL-6, and VEGF-C levels in septic mice as well as inhibit phosphorylation of p38 and ERK1/2 in lung tissues of septic mice. This is the first study demonstrating the protective effect of endostatin on sepsis and its possible underlying mechanisms from the aspects of inhibiting inflammatory responses, blocking VEGF receptor, attenuating VEGF-C expression, and reducing vascular permeability. Overall, the study revealed the potential protect role for endostatin in the treatment of sepsis.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Endostatinas/uso terapêutico , Insuficiência de Múltiplos Órgãos/prevenção & controle , Sepse/complicações , Inibidores da Angiogênese/administração & dosagem , Animais , Citocinas/biossíntese , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Endostatinas/administração & dosagem , Mediadores da Inflamação/sangue , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Insuficiência de Múltiplos Órgãos/metabolismo , Insuficiência de Múltiplos Órgãos/microbiologia , Sepse/metabolismo , Análise de Sobrevida , Fator A de Crescimento do Endotélio Vascular/sangue
16.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 30(5): 525-8, 2005 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-16320580

RESUMO

OBJECTIVE: To construct the hepatitis B virus (HBV) X gene recombinant and to induce the expression of X protein. METHODS: HBV DNA was extracted from the serum of patient with hepatitis B. The X gene was amplified by PCR using the primers with EcoRI and HindIII digestion sites, and then cloned into pronucleus expression vector pMAL-C2X, which was detected by EcoRI and HindIII digestion and sequence. Finally, the recombinant was induced by IPTG to express X protein in JM109. RESULTS: The band similar to X gene was amplified by PCR. There were fragments similar to X gene when the recombinant was digested by the enzyme digestion. It was tested by DNA sequence that the correct and entire opening reading frame of HBV X gene was inserted. The X protein was expressed by the IPTG induction. CONCLUSION: Pronucleus expression recombinant pMAL-C2X-HBV-X is constructed successfully and with the IPTG induction, the recombinant pMAL-C2X-HBV-X can express the X protein in E. coli JM109, which lays the foundation for the HBV X protein purification and its biological study.


Assuntos
Vírus da Hepatite B/genética , Transativadores/biossíntese , Transfecção , Sequência de Bases , Clonagem Molecular , Escherichia coli/genética , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Fases de Leitura Aberta/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Análise de Sequência de DNA , Transativadores/genética , Proteínas Virais Reguladoras e Acessórias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA