Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
J Surg Res ; 256: 584-594, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32805581

RESUMO

BACKGROUND: Acute lung injury (ALI), manifested by progressive hypoxemia and respiratory distress, is associated with high morbidity and mortality, which lacks the effective therapies in clinics. Our previous studies demonstrated that maresin1 (MaR1), a specialized proresolving mediator, could effectively mitigate the inflammation of lipopolysaccharide (LPS)-induced ALI. However, whether MaR1 impacts the macrophage polarization to alleviate ALI remains unclear. Our study explored the effects and underlying mechanisms of MaR1 on the macrophage phenotypes in ALI. MATERIAL AND METHODS: Male BALB/c mice were subjected to endotracheal instillation of LPS to induce ALI and then intravenously injected with MaR1 or normal saline. Intraperitoneal administration of peroxisome proliferator-activated receptor-γ (PPAR-γ) inhibitor GW9662 was given 30 mins before MaR1. We measured the pathohistologic changes, pulmonary edema, inflammatory cytokines, and the flow cytometry of macrophage phenotypes. RESULTS: Our results illustrated that MaR1 ameliorated lung injury and increased monocyte or macrophage recruitment and the release of anti-inflammatory cytokines. The flow cytometry showed that MaR1 promoted polarization of CD11c-CD206+ (M2) macrophages and inhibited polarization of CD11c+CD206- (M1) macrophages. Besides, the western blotting revealed that MaR1 increased the expression of PPAR-γ. The pretreatment with PPAR-γ antagonist GW9662 could significantly suppress the polarization of M2 macrophages and antagonize the protective effects of MaR1 on LPS-stimulated ALI. CONCLUSIONS: MaR1 was able to promote M2 macrophage polarization by reversing LPS-mediated PPAR-γ inhibition, thereby expediting the recovery of LPS-stimulated ALI.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Ácidos Docosa-Hexaenoicos/farmacologia , Macrófagos/efeitos dos fármacos , PPAR gama/agonistas , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Anilidas/administração & dosagem , Animais , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/uso terapêutico , Humanos , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , PPAR gama/antagonistas & inibidores , PPAR gama/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
2.
Mediators Inflamm ; 2020: 2431705, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32317861

RESUMO

CXCL14 is a relatively novel chemokine with a wide spectrum of biological activities. The present study was designed to investigate whether CXCL14 overexpression attenuates sepsis-associated acute kidney injury (AKI) in mice. Sepsis model has been established by cecal ligation and puncture (CLP). CLP induced AKI in mice as assessed by increased renal neutrophil gelatinase-associated lipocalin (NGAL) expression and serum creatinine levels. We found that renal CXCL14 expression in the kidney was significantly decreased at 12 hours after CLP. Correlation analysis demonstrated a negative association between renal CXCL14 expression and AKI markers including serum creatinine and renal NGAL. Moreover, CXCL14 overexpression reduced cytokine (TNF-α, IL-6, and IL-1ß) production and NGAL expression in the kidney and decreased serum creatinine levels. In vivo and in vitro experiments found that CXCL14 overexpression inhibited M1 macrophage polarization but increased M2 polarization. Together, these results suggest that CXCL14 overexpression attenuates sepsis-associated AKI probably through the downregulation of macrophages-derived cytokine production. However, further studies are required to elucidate the underlying mechanism.


Assuntos
Quimiocinas CXC/metabolismo , Sepse/metabolismo , Animais , Western Blotting , Quimiocinas CXC/genética , Creatinina/metabolismo , Ensaio de Imunoadsorção Enzimática , Rim/metabolismo , Rim/patologia , Lentivirus/genética , Ligadura/efeitos adversos , Lipocalina-2/genética , Lipocalina-2/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Punções/efeitos adversos , Células RAW 264.7 , Reação em Cadeia da Polimerase em Tempo Real , Sepse/genética
3.
Lab Invest ; 99(8): 1143-1156, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30911150

RESUMO

Acute respiratory distress syndrome (ARDS) is a uniform progression of overwhelming inflammation in lung tissue with extensive infiltration of inflammatory cells. Neutrophil apoptosis is thought to be a significant process in the control of the resolution phase of inflammation. It has been proved that 5-Aza-2'-deoxycytidine (Aza) can inhibit cancer by activating death-associated protein kinase 1 (DAPK1) to promote apoptosis. However, the effect of DAPK1 on neutrophil apoptosis is unclear, and research on the role of Aza in inflammation is lacking. Here, we investigated whether Aza can regulate DAPK1 expression to influence the fate of neutrophils in ARDS. In vitro, we stimulated neutrophil-like HL-60 (dHL-60) cells with different concentrations of Aza for different durations and used RNA interference to up- or downregulate DAPK1 expression. We observed that culturing dHL-60 cells with Aza increased apoptosis by inhibiting NF-κB activation to modulate the expression of Bcl-2 family proteins, which was closely related to the levels of DAPK1. In vivo, ARDS was evoked by intratracheal instillation of lipopolysaccharide (LPS; 3 mg/kg). One hour after LPS administration, mice were treated with Aza (1 mg/kg, i.p.). To inhibit DAPK1 expression, mice were intraperitoneally injected with a DAPK1 inhibitor. Aza treatment accelerated inflammatory resolution in LPS-induced ARDS by suppressing pulmonary edema, alleviating lung injury and decreasing the infiltration of inflammatory cells in bronchoalveolar lavage fluid (BALF). Moreover, Aza reduced the production of proinflammatory cytokines. However, administration of the DAPK1 inhibitor attenuated the protective effects of Aza. Similarly, the proapoptotic function of Aza was prevented when DAPK1 was inhibited either in vivo or in vitro. In summary, Aza promotes neutrophil apoptosis by activating DAPK1 to accelerate inflammatory resolution in LPS-induced ARDS. This study provides the first evidence that Aza prevents LPS-induced neutrophil survival by modulating DAPK1 expression.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas Quinases Associadas com Morte Celular , Inflamação/metabolismo , Neutrófilos/efeitos dos fármacos , Síndrome do Desconforto Respiratório/metabolismo , Animais , Citocinas/metabolismo , Proteínas Quinases Associadas com Morte Celular/metabolismo , Proteínas Quinases Associadas com Morte Celular/farmacologia , Decitabina/metabolismo , Decitabina/farmacologia , Modelos Animais de Doenças , Células HL-60 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
4.
J Huazhong Univ Sci Technolog Med Sci ; 35(2): 225-229, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25877356

RESUMO

The possible role of minocycline in microglial activation and neuronal death after cardiac arrest (CA) and cardiopulmonary resuscitation (CPR) in mice was investigated in this study. The mice were given potassium chloride to stop the heart beating for 8 min to achieve CA, and they were subsequently resuscitated with epinephrine and chest compressions. Forty adult C57BL/6 male mice were divided into 4 groups (n=10 each): sham-operated group, CA/CPR group, CA/CPR+minocycline group, and CA/CPR+vehicle group. Animals in the latter two groups were intraperitoneally injected with minocycline (50 mg/kg) or vehicle (normal saline) 30 min after recovery of spontaneous circulation (ROSC). Twenty-four h after CA/CPR, the brains were removed for histological evaluation of the hippocampus. Microglial activation was evaluated by detecting the expression of ionized calcium-binding adapter molecule-1 (Iba1) by immunohistochemistry. Neuronal death was analyzed by hematoxylin and eosin (H&E) staining and the levels of tumor necrosis factor-alpha (TNF-α) in the hippocampus were measured by enzyme-linked immunosorbent assay (ELISA). The results showed that the neuronal death was aggravated, most microglia were activated and TNF-α levels were enhanced in the hippocampus CA1 region of mice subjected to CA/CPR as compared with those in the sham-operated group (P<0.05). Administration with minocycline 30 min after ROSC could significantly decrease the microglial response, TNF-α levels and neuronal death (P<0.05). It was concluded that early administration with minocycline has a strong therapeutic potential for CA/CPR-induced brain injury.


Assuntos
Reanimação Cardiopulmonar , Morte Celular/efeitos dos fármacos , Parada Cardíaca/patologia , Microglia/efeitos dos fármacos , Minociclina/farmacologia , Neurônios/efeitos dos fármacos , Animais , Ensaio de Imunoadsorção Enzimática , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/citologia , Fator de Necrose Tumoral alfa/metabolismo
5.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 37(5): 541-8, 2015 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-26564505

RESUMO

OBJECTIVE: To identify the incidence and risk factors of perioperative major adverse cardiac events (MACE) in elderly patients with coronary heart disease (CHD) undergoing non-cardiac surgery. METHODS: We prospectively analyzed the clinical data of 360 CHD patients who aged 75 years or older undergoing elective intermediate-to high-risk surgery in five medical centers across China from January 2008 to January 2010. The clinical variables included the 12-lead ECG and Troponin I levels after surgery. The combined outcome was defined as all the perioperative MACE in hospital. The risk factors of MACE and their indexes were analyzed with univariate analysis and multivariable logistic regression in SPSS software,together with a risk scoring and stratification system established. RESULTS: Perioperative MACE occurred in 11.94% of elderly CHD patients undergoing non-cardiac surgery. Seven independent risk factors of perioperative MACE for this population were identified,which included angina within 6 months (P=0.001), hypertension(P=0.014), preoperative haematocrit (HCT) <40% (P=0.050), serum creatinine (Scr)>150 mmol/L (P=0.014), ejection fraction(EF) <50% (P=0.019), intraoperative hyoxemia (P=0.019), and operative time>150 min (P=0.001). The risk indexes of these factors were 4,3,3,6,4,5, and 4, respectively. The rate of perioperative MACE increased significantly as the level of risk stratification elevated. CONCLUSIONS: Elderly CHD patients undergoing non-cardiac surgery are at high risk of perioperative MACE. Angina within 6 months,hypertension, preoperative HCT<40%, Scr>150 mmol/L, EF<50%, intraoperative hyoxemia, and operative time>150 min can increase the risk of MACE. The risk scoring and stratification system based on the risk factor index can be a valuable parameter for assessing the perioperative cardiac risk of noncardiac surgery for elderly CHD patients.


Assuntos
Doença das Coronárias , Idoso , China , Procedimentos Cirúrgicos Eletivos , Eletrocardiografia , Humanos , Incidência , Modelos Logísticos , Assistência Perioperatória , Fatores de Risco
6.
J Surg Res ; 183(2): 710-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23558258

RESUMO

BACKGROUND: Hemorrhagic shock activates cellular stress signals and can lead to systemic inflammatory response, organ injury, and death. Mitogen-activated protein kinase (MAPK) acts as a sensor of tissue injury in models of ischemia-reperfusion injury. Lipoxins are endogenous lipid mediators with potent anti-inflammatory and pro-resolving actions. We hypothesized that BML-111 (a lipoxin A4-receptor agonist) attenuates hemorrhagic shock-induced acute lung injury (ALI) through inhibiting activation of the MAPK pathway. METHODS: We randomized Sprague-Dawley rats into four groups: sham, hemorrhagic shock-resuscitation (HS), HS plus BML-111 (BML-111), and HS plus BML-111 and BOC-2 (BOC-2). Two hours after resuscitation, we collected samples of lung. We obtained bronchoalveolar lavage fluid for neutrophil count. We performed optical microscopy to examine pathologic changes in lungs. Wet/dry ratios, myeloperoxidase expression, interleukin (IL)-1ß and IL-6 levels in lung were measured. We evaluated MAPK activation and the DNA binding activity of activator protein-1 in lung. RESULTS: Treatment with BML-111 reduced the lung damage and wet/dry ratio, neutrophil count in bronchoalveolar lavage fluid, expression of myeloperoxidase, and production of IL-1ß and IL-6 in lung. Phosphorylation of MAPK was also decreased by BML-111 in lung. Furthermore, the DNA binding activity of activator protein-1 was blocked by BML-111. An antagonist of the lipoxin A4-receptor, BOC-2, reversed the protective effect of BML-111 on ALI induced by hemorrhagic shock. CONCLUSIONS: This study indicates that BML-111 attenuated hemorrhagic shock-induced ALI via the MAPK/activator protein-1 signaling pathway. Therefore, BML-111 may have therapeutic potential for hemorrhagic shock-induced ALI.


Assuntos
Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/prevenção & controle , Ácidos Heptanoicos/uso terapêutico , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Choque Hemorrágico/complicações , Transdução de Sinais/fisiologia , Lesão Pulmonar Aguda/metabolismo , Animais , Modelos Animais de Doenças , Ácidos Heptanoicos/farmacologia , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Oligopeptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Lipoxinas/agonistas , Receptores de Lipoxinas/antagonistas & inibidores , Receptores de Lipoxinas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição AP-1/antagonistas & inibidores , Fator de Transcrição AP-1/efeitos dos fármacos , Fator de Transcrição AP-1/fisiologia
7.
Mil Med Res ; 9(1): 58, 2022 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-36229865

RESUMO

BACKGROUND: Abnormal myocardial Nav1.5 expression and function cause lethal ventricular arrhythmias during myocardial ischemia-reperfusion (I/R). Protein inhibitor of activated STAT Y (PIASy)-mediated caveolin-3 (Cav-3) SUMO modification affects Cav-3 binding to the voltage-gated sodium channel 1.5 (Nav1.5). PIASy activity is increased after myocardial I/R, but it is unclear whether this is attributable to plasma membrane Nav1.5 downregulation and ventricular arrhythmias. METHODS: Using recombinant adeno-associated virus subtype 9 (AAV9), rat cardiac PIASy was silenced using intraventricular injection of PIASy short hairpin RNA (shRNA). After two weeks, rat hearts were subjected to I/R and electrocardiography was performed to assess malignant arrhythmias. Tissues from peri-infarct areas of the left ventricle were collected for molecular biological measurements. RESULTS: PIASy was upregulated by I/R (P < 0.01), with increased SUMO2/3 modification of Cav-3 and reduced membrane Nav1.5 density (P < 0.01). AAV9-PIASy shRNA intraventricular injection into the rat heart downregulated PIASy after I/R, at both mRNA and protein levels (P < 0.05 vs. Scramble-shRNA + I/R group), decreased SUMO-modified Cav-3 levels, enhanced Cav-3 binding to Nav1.5, and prevented I/R-induced decrease of Nav1.5 and Cav-3 co-localization in the intercalated disc and lateral membrane. PIASy silencing in rat hearts reduced I/R-induced fatal arrhythmias, which was reflected by a modest decrease in the duration of ventricular fibrillation (VF; P < 0.05 vs. Scramble-shRNA + I/R group) and a significantly reduced arrhythmia score (P < 0.01 vs. Scramble-shRNA + I/R group). The anti-arrhythmic effects of PIASy silencing were also evidenced by decreased episodes of ventricular tachycardia (VT), sustained VT and VF, especially at the time 5-10 min after ischemia (P < 0.05 vs. Scramble-shRNA + IR group). Using in vitro human embryonic kidney 293 T (HEK293T) cells and isolated adult rat cardiomyocyte models exposed to hypoxia/reoxygenation (H/R), we confirmed that increased PIASy promoted Cav-3 modification by SUMO2/3 and Nav1.5/Cav-3 dissociation after H/R. Mutation of SUMO consensus lysine sites in Cav-3 (K38R or K144R) altered the membrane expression levels of Nav1.5 and Cav-3 before and after H/R in HEK293T cells. CONCLUSIONS: I/R-induced cardiac PIASy activation increased Cav-3 SUMOylation by SUMO2/3 and dysregulated Nav1.5-related ventricular arrhythmias. Cardiac-targeted PIASy silencing mediated Cav-3 deSUMOylation and partially prevented I/R-induced Nav1.5 downregulation in the plasma membrane of cardiomyocytes, and subsequent ventricular arrhythmias in rats. PIASy was identified as a potential therapeutic target for life-threatening arrhythmias in patients with ischemic heart diseases.


Assuntos
Antiarrítmicos , Caveolina 3 , Proteínas de Ligação a Poli-ADP-Ribose/genética , Proteínas Inibidoras de STAT Ativados/genética , Animais , Arritmias Cardíacas/genética , Caveolina 3/genética , Caveolina 3/metabolismo , Regulação para Baixo , Inativação Gênica , Células HEK293 , Humanos , Isquemia/complicações , Lisina/genética , Lisina/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , RNA Mensageiro , RNA Interferente Pequeno , Ratos , Reperfusão/efeitos adversos
8.
J Neuroinflammation ; 8: 95, 2011 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-21831303

RESUMO

BACKGROUND: Microglial activation plays an important role in neurodegenerative diseases through production of nitric oxide (NO) and several pro-inflammatory cytokines. Lipoxins (LXs) and aspirin-triggered LXs (ATLs) are considered to act as 'braking signals' in inflammation. In the present study, we investigated the effect of aspirin-triggered LXA4 (ATL) on infiammatory responses induced by lipopolysaccharide (LPS) in murine microglial BV-2 cells. METHODS: BV-2 cells were treated with ATL prior to LPS exposure, and the effects of such treatment production of nitric oxide (NO), inducible nitric oxide synthase (iNOS), interleukin-1ß (IL-1ß) and tumour necrosis factor-α (TNF-α) were analysed by Griess reaction, ELISA, western blotting and quantitative RT-PCR. Moreover, we investigated the effects of ATL on LPS-induced nuclear factor-κB (NF-κB) activation, phosphorylation of mitogen-activated protein kinases (MAPKs) and activator protein-1 (AP-1) activation. RESULTS: ATL inhibited LPS-induced production of NO, IL-1ß and TNF-α in a concentration-dependent manner. mRNA expressions for iNOS, IL-1ß and TNF-α in response to LPS were also decreased by ATL. These effects were inhibited by Boc-2 (a LXA4 receptor antagonist). ATL significantly reduced nuclear translocation of NF-κB p65, degradation of the inhibitor IκB-α, and phosphorylation of extracellular signal-regulated kinase (ERK) and p38 MAPK in BV-2 cells activated with LPS. Furthermore, the DNA binding activity of NF-κB and AP-1 was blocked by ATL. CONCLUSIONS: This study indicates that ATL inhibits NO and pro-inflammatory cytokine production at least in part via NF-κB, ERK, p38 MAPK and AP-1 signaling pathways in LPS-activated microglia. Therefore, ATL may have therapeutic potential for various neurodegenerative diseases.


Assuntos
Aspirina/farmacologia , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Lipoxinas/farmacologia , Microglia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Linhagem Celular , Ativação Enzimática , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipoxinas/metabolismo , Camundongos , Microglia/citologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Proteínas Quinases Ativadas por Mitógeno/genética , NF-kappa B/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
9.
Curr Med Sci ; 41(5): 974-980, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34562209

RESUMO

OBJECTIVE: To evaluate the analgesic effect of ultrasound-guided subcostal anterior quadratus lumborum block (QLB) for laparoscopic radical gastrectomy surgery. METHODS: Patients (aged 20-65 years, ASA I - II, and weighing 40-75 kg) scheduled for elective laparoscopic radical gastrectomy were enrolled in the current study. Sixty patients were randomly assigned to two groups by computer-generated randomization codes: an ultrasound-guided oblique subcostal transversus abdominis plane block (TAPB) group (group T, n=30) or an ultrasound-guided subcostal anterior QLB group (group Q, n=30). In both groups, bilateral ultrasound-guided oblique subcostal TAPB and subcostal anterior QLB were performed before general anesthesia with 0.25% ropivacaine 0.5 mL/kg. For postoperative management, all patients received patient-controlled intravenous analgesia (PCIA) with nalbuphine and sufentanil after surgery, maintaining visual analogue scale (VAS) scores ≤4 within 48 h. The intraoperative consumption of remifentanil, the requirement for sufentanil as a rescue analgesic, and the VAS scores at rest and coughing were recorded at 1, 6, 12, 24 and 48 h after surgery. The recovery (extubation time after surgery, first ambulation time, first flatus time and length of postoperative hospital stay) and the adverse events (nausea and vomiting, skin pruritus, respiratory depression and nerve-block related complications) were observed and recorded. The primary outcome was the perioperative consumption of opioids. RESULTS: Compared with group T, the intraoperative consumption of remifentanil, requirement for sufentanil and the frequency of PCIA were reduced in group Q. Meanwhile, VAS scores at all points of observation were significantly lower in group Q than in group T. Patients in group Q were also associated with shorter time to first out-of-bed activity and flatus, and shorter length of postoperative hospital stay than group T (P<0.05). There were no skin pruritus, respiratory depression or nerve-block related complications in both groups. CONCLUSION: Compared with ultrasound-guided oblique subcostal TAPB, ultrasound-guided subcostal anterior QLB provided greater opioid-sparing effect, lower visual analogue scores, and shorter postoperative hospital stay for laparoscopic radical gastrectomy.


Assuntos
Músculos Abdominais/diagnóstico por imagem , Gastrectomia/métodos , Nalbufina/administração & dosagem , Dor Pós-Operatória/tratamento farmacológico , Remifentanil/administração & dosagem , Ropivacaina/administração & dosagem , Sufentanil/administração & dosagem , Adulto , Anestesia Geral , Feminino , Humanos , Laparoscopia , Masculino , Pessoa de Meia-Idade , Bloqueio Nervoso/métodos , Medição da Dor , Distribuição Aleatória , Ultrassonografia de Intervenção , Adulto Jovem
10.
Chin Med J (Engl) ; 133(5): 590-596, 2020 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-32032080

RESUMO

Enhanced recovery after cesarean (ERAC) delivery is an evidence-based, multi-disciplinary approach throughout pre-, intra-, post-operative period. The ultimate goal of ERAC is to enhance recovery and improve the maternal and neonatal outcomes. This review highlights the role of anesthesiologist in ERAC protocols. This review provided a general introduction of ERAC including the purposes and the essential elements of ERAC protocols. The tool used for evaluating the quality of ERAC (ObsQoR-11) was discussed. The role of anesthesiologist in ERAC should cover the areas including management of peri-operative hypotension, prevention and treatment of intra- and post-operative nausea and vomiting, prevention of hypothermia and multi-modal peri-operative pain management, and active pre-operative management of unplanned conversion of labor analgesia to cesarean delivery anesthesia. Although some concerns still remain, ERAC implementation should not be delayed. Regular assessment and process improvement should be imbedded into the protocol. Further high-quality studies are warranted to demonstrate the effectiveness and efficacy of the ERAC protocol.


Assuntos
Cesárea/métodos , Recuperação Pós-Cirúrgica Melhorada , Anestesiologistas , Feminino , Humanos , Náusea e Vômito Pós-Operatórios , Gravidez
11.
Chin Med J (Engl) ; 133(2): 183-189, 2020 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-31929368

RESUMO

BACKGROUND: Obstetric hemorrhage is a major cause of maternal death during cesarean delivery. The objective of this retrospective observational study was to evaluate the efficacy and safety of intra-operative cell salvage (IOCS) in cesarean section. METHODS: We included a total of 361 patients diagnosed with central placenta previa who underwent cesarean section from May 2016 to December 2018. In this study, 196 patients received autologous transfusion using IOCS (IOCS group) and 165 patients accepted allogeneic blood transfusion (ABT group). Propensity score matched analysis was performed to balance differences in the baseline variables between the IOCS group and ABT group. Patients in the IOCS group were matched 1:1 to patients in the ABT group. RESULTS: After propensity score matching, 137 pairs of cases between the two groups were successfully matched and no significant differences in baseline characteristics were found between the IOCS group and ABT group. Patients in the IOCS group were associated with significantly shorter length of hospital stay, compared with ABT group (8.9 ±â€Š4.1 days vs. 10.3 ±â€Š5.2 days, t = -2.506, P = 0.013). The postoperative length of hospital stay was 5.3 ±â€Š1.4 days for patients in the IOCS group and 6.6 ±â€Š3.6 days for those in the ABT group (t = -4.056, P < 0.001). The post-operative hemoglobin level in the IOCS group and ABT group was 101.3 ±â€Š15.4 and 96.3 ±â€Š16.6 g/L, respectively, which were significantly different (t = 2.615, P = 0.009). Allogeneic red blood cell transfusion was significantly lower at 0 unit (range: 0-11.5 units) in the IOCS group when compared with 2 units (range: 1-20 units) in the ABT group (P < 0.001). CONCLUSIONS: This retrospective observational study using propensity score matched analysis suggested that IOCS was associated with shorter length of postoperative hospital stay and higher post-operative hemoglobin levels during cesarean delivery.


Assuntos
Cesárea/estatística & dados numéricos , Adulto , Transfusão de Sangue/métodos , Feminino , Hemoglobinas/metabolismo , Humanos , Tempo de Internação , Período Pós-Operatório , Gravidez , Pontuação de Propensão , Estudos Retrospectivos
12.
Curr Med Sci ; 40(4): 773-785, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32862390

RESUMO

Renal cancer is a common genitourinary malignance, of which clear cell renal cell carcinoma (ccRCC) has high aggressiveness and leads to most cancer-related deaths. Identification of sensitive and reliable biomarkers for predicting tumorigenesis and progression has great significance in guiding the diagnosis and treatment of ccRCC. Here, we identified 2397 common differentially expressed genes (DEGs) using paired normal and tumor ccRCC tissues from GSE53757 and The Cancer Genome Atlas (TCGA). Then, we performed weighted gene co-expression network analysis and protein-protein interaction network analysis, 17 candidate hub genes were identified. These candidate hub genes were further validated in GSE36895 and Oncomine database and 14 real hub genes were identified. All the hub genes were up-regulated and significantly positively correlated with pathological stage and histologic grade of ccRCC. Survival analysis showed that the higher expression level of each hub gene tended to predict a worse clinical outcome. ROC analysis showed that all the hub genes can accurately distinguish between tumor and normal samples, and between early stage and advanced stage ccRCC. Moreover, all the hub genes were positively associated with distant metastasis, lymph node infiltration, tumor recurrence and the expression of MKi67, suggesting these genes might promote tumor proliferation, invasion and metastasis. Furthermore, the functional annotation demonstrated that most genes were enriched in cell-cycle related biological function. In summary, our study identified 14 potential biomarkers for predicting tumorigenesis and progression, which might contribute to early diagnosis, prognosis prediction and therapeutic intervention.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma de Células Renais/diagnóstico , Perfilação da Expressão Gênica/métodos , Neoplasias Renais/diagnóstico , Regulação para Cima , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Progressão da Doença , Detecção Precoce de Câncer , Feminino , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , Mapas de Interação de Proteínas , Análise de Sobrevida
13.
Int Immunopharmacol ; 81: 106257, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32044659

RESUMO

The activation of NLRP3 inflammasome and NF-κB pathway, associating with oxidativestress, have been implicated in the development of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). NecroX-5 has been reported to exhibit theeffectsofanti-oxidation and anti-stress in various diseases. However, the role of NecroX-5 in ALI has not been explicitly demonstrated. The aim of this study was to explore the therapeutic effects and potential mechanism action of NecroX-5 on ALI. Here, we found that NecroX-5 pretreatment dramatically diminished the levels of IL-1ß, IL-18 and ROS in in RAW264.7 cells challenged with LPS and ATP. Furthermore, NecroX-5 suppressed the activation of NLRP3 inflammasome and NF-κB signalpathway. In addition, NecroX-5 also inhibited the thioredoxin-interacting protein (TXNIP) expression. In vivo, NecroX-5 reduced the LPS-induced lung histopathological injury, the number of TUNEL-positive cells, lung wet/dry (W/D) ratio, levels of total protein and inflammatory cytokines in the bronchoalveolar lavage fluid (BALF) in mice. Additionally, LPS-induced upregulation of myeloperoxidase (MPO), ROS production and malondialdehyde (MDA) were inhibited by NecroX-5 administration. Thus, our results demonstrate that NecroX-5 protects against LPS-induced ALI by inhibiting TXNIP/NLRP3 and NF-κB.


Assuntos
Anti-Inflamatórios/uso terapêutico , Proteínas de Transporte/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/uso terapêutico , Pulmão/metabolismo , Síndrome do Desconforto Respiratório/tratamento farmacológico , Sulfonas/uso terapêutico , Tiorredoxinas/metabolismo , Animais , Proteínas de Transporte/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Lipopolissacarídeos/imunologia , Pulmão/patologia , Masculino , Camundongos , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Peroxidase/metabolismo , Células RAW 264.7 , Síndrome do Desconforto Respiratório/imunologia , Transdução de Sinais , Tiorredoxinas/genética
14.
Exp Biol Med (Maywood) ; 233(2): 200-8, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18222975

RESUMO

Cardiomyocyte hypertrophy is formed in response to pressure or volume overload, injury, or neurohormonal activation. The most important vascular hormone that contributes to the development of hypertrophy is angiotensin II (Ang II). Accumulating studies have suggested that reactive oxygen species (ROS) may play an important role in cardiac hypertrophy. Propofol is a general anesthetic that possesses antioxidant action. We therefore examined whether propofol inhibited Ang II-induced cardiomyocyte hypertrophy. Our results showed that both ROS formation and hypertrophic responses induced by Ang II in cardiomyocytes were partially blocked by propofol. Further studies showed that propofol inhibited the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) and mitogen-activated protein kinase/ERK kinase 1/2 (MEK1/2) induced by Ang II via a decrease in ROS production. In addition, propofol also markedly attenuated Ang II-stimulated nuclear factor-kappaB (NF-kappaB) activation via a decrease in ROS production. In conclusion, propofol prevents cardiomyocyte hypertrophy by interfering with the generation of ROS and involves the inhibition of the MEK/ERK signaling transduction pathway and NF-kappaB activation.


Assuntos
Angiotensina II/farmacologia , Cardiomegalia , Miócitos Cardíacos/efeitos dos fármacos , Propofol/farmacologia , Animais , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citoproteção/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Miócitos Cardíacos/metabolismo , NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
15.
Crit Care ; 12(1): R5, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18199317

RESUMO

INTRODUCTION: Fungal infection is increasingly common in critical illness with severe sepsis, but the influence of invasive fungal infection (IFI) on severe sepsis is not well understood. The aim of this study was to investigate the impact that IFI has on the outcomes of critically ill surgical patients with severe sepsis in China by means of matched cohort analysis; we also evaluated the epidemiologic characteristics of IFI in this population. METHODS: Records for all admissions to 10 university hospital surgical intensive care units (ICUs) from December 2004 to November 2005 were reviewed. Patients who met criteria for severe sepsis were included. IFI was identified using established criteria based on microbiologic or histological evidence. A matched cohort study was conducted to analyze the relationship between IFI and outcomes of severe sepsis. RESULTS: A total of 318 patients with severe sepsis were enrolled during the study period, of whom 90 (28.3%) were identified as having IFI. A total of 100 strains of fungi (58% Candida albicans) were isolated from these patients. Independent risk factors for IFI in patients with severe sepsis included mechanical ventilation (>3 days), Acute Physiology and Chronic Health Evaluation score, coexisting infection with both gram-positive and gram-negative bacteria, and urethral catheterization (>3 days). Compared with the control cohort, IFI was associated with increased hospital mortality (P < 0.001), high hospital costs (P = 0.038), and prolonged stay in the ICU (P < 0.001) and hospital (P = 0.020). CONCLUSION: IFI is frequent in patients with severe sepsis in surgical ICUs and is associated with excess risk for hospital mortality, longer ICU and hospital stays, and greater consumption of medical resources.


Assuntos
Mortalidade Hospitalar , Micoses/complicações , Complicações Pós-Operatórias , Sepse/complicações , APACHE , Idoso , Estudos de Coortes , Feminino , Humanos , Unidades de Terapia Intensiva , Modelos Logísticos , Masculino , Prontuários Médicos , Pessoa de Meia-Idade , Micoses/classificação , Fatores de Risco , Sepse/classificação , Sepse/terapia , Índice de Gravidade de Doença
16.
Chin Med J (Engl) ; 121(1): 49-55, 2008 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-18208666

RESUMO

BACKGROUND: Urinary trypsin inhibitor inhibits the enhanced production of pro-inflammatory molecules. Hemeoxygenase-1 induction protects against ischemia/reperfusion injury, oxidative stress, inflammation, transplant rejection, apoptosis, and other conditions. However, it is unknown if a combined hemin and ulinastatin pretreatment could result in protective effects for septic shock. In this study, we investigated the role of hemin pretreatment combined with ulinastatin on septic shock in rats. METHODS: Eighty healthy, male Sprague-Dawley rats were randomly divided into four groups: group S, group H, group U and group HU. Groups S and U received 1 ml normal saline intraperitoneally, while groups H and HU both received 1 ml (100 mg /kg) hemin. Twenty-four hours later, 0.5 ml (10 mg/kg) E. coli lipopolysaccharide was injected intravenously to replicate the experimental model of septic shock. After an initial 25% decrease in the mean arterial pressure, corresponding to time point 0, groups HU and U received 0.5 ml 10 000 U/kg ulinastatin intravenously, and the others received 0.5 ml normal saline. RESULTS: The number of deaths in groups H and U was lower than that in the group S (P < 0.05), and was higher than that in group HU (all P < 0.05) respectively. The mean arterial pressure (MAP) in the group S was significantly greater than that in group H (P < 0.05), and was lower than that in group HU and group U (P < 0.05). The plasma levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), creatinine (Cr) and blood urea nitrogen (BUN), the malondial-dehyde (MDA) of liver, kidney and lung, and the lung Evans blue (EB) contents in groups H and U, were greater than that in group HU (all P < 0.05), and were lower than that in group S (all P < 0.05). In contrast, the plasma levels of CO in groups H and HU were higher than that in groups S and U (all P < 0.05), and SOD of liver, kidney and lung in groups H and U were higher than that in group S, and were lower than that in group HU (all P < 0.05). The levels of TNF-alpha, IL-6, IL-8 and beta-glucuronidase (GCD) activity of plasma in groups U and HU were lower than those in groups H and S, all having a P < 0.05, while there were no significant differences between group H and group S, or between group HU and group U (all P > 0.05). The HO-1 mRNA and HO-1 protein levels from hepatic, renal, and pulmonary tissue in groups S and U were lower than those in groups H and HU (all P < 0.05), but there were no significant differences between groups S and U, or between groups H and HU (all P > 0.05). The HO-2 mRNA and HO-2 protein were not significantly different among the four groups (all P > 0.05). CONCLUSIONS: Combined pretreatment with hemin and ulinastatin in septic shock rats results in an improved response by the upregulation of HO-1 protein followed by increasing CO with resistance to increased oxidative stress, restraining the release of inflammatory mediators, and inhibiting beta-GCD activity.


Assuntos
Glicoproteínas/uso terapêutico , Hemina/uso terapêutico , Choque Séptico/tratamento farmacológico , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Pressão Sanguínea/efeitos dos fármacos , Nitrogênio da Ureia Sanguínea , Creatinina/sangue , Citocinas/sangue , Heme Oxigenase (Desciclizante)/análise , Heme Oxigenase (Desciclizante)/genética , Masculino , Malondialdeído/sangue , Ratos , Ratos Sprague-Dawley , Choque Séptico/fisiopatologia , Superóxido Dismutase/metabolismo
17.
Chin Med J (Engl) ; 121(4): 341-6, 2008 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-18304468

RESUMO

BACKGROUND: Isoflurane, a commonly used inhaled anesthetic, induces apoptosis in primary rat cortical neurons of rat in a concentration- and time-dependent manner by an unknown mechanism. We hypothesized that isoflurane induced apoptosis by causing abnormal calcium release from the endoplasmic reticulum (ER) via activation of inositol 1, 4, 5-trisphosphate (IP(3)) receptors. Sevoflurane has a reduced ability to disrupt intracellular calcium homeostasis and is a less potent cytotoxic agent. This study examined and compared the cytotoxic effects of isoflurane and sevoflurane on rat primary cortical neurons and their relationship with disruption of intracellular calcium homeostasis and production of reactive oxygen species (ROS). METHODS: Primary rat cortical neurons were treated with the equivalent of 1 minimal alveolar concentration (MAC) of isoflurane and sevoflurane for 12 hours. MTT reduction and LDH release assays were performed to evaluate cell viability. Changes of calcium concentration in the cytosolic space, [Ca(2+)](c), and production of ROS were determined after exposing primary rat cortical neurons to isoflurane and sevoflurane. We also determined the effects of IP(3) receptor antagonist xestospongin C on isoflurane-induced cytotoxicity and calcium release from the ER in primary rat cortical neurons. RESULTS: Isoflurane at 1 MAC for 12 hours induced cytotoxicity in primary rat cortical neurons, which was also associated with a high and fast elevation of peak [Ca(2+)](c). Xestospongin C significantly ameliorated isoflurane cytotoxicity in primary cortical neurons, as well as inhibited the calcium release from the ER in primary cortical neurons. Isoflurane did not induce significant changes of ROS production in primary rat cortical neurons. Sevoflurane, at equivalent exposure to isoflurane, did not induce similar cytotoxicity or elevation of peak [Ca(2+)](c) in primary rat cortical neurons. CONCLUSION: These results suggested that isoflurane induced elevation in [Ca(2+)](c), partially via elevated activity of IP(3) receptors, which rendered cells vulnerable to isoflurane neurotoxicity. ROS production was not involved in isoflurane-induced neurotoxicity. Sevoflurane, at an equivalent exposure to isoflurane, did not induce similar elevations of [Ca(2+)](c) or neurotoxicity in primary cortical neurons of rat.


Assuntos
Anestésicos Inalatórios/toxicidade , Isoflurano/toxicidade , Éteres Metílicos/toxicidade , Animais , Cálcio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Receptores de Inositol 1,4,5-Trifosfato/efeitos dos fármacos , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Sevoflurano
18.
Zhonghua Yi Xue Za Zhi ; 88(13): 876-9, 2008 Apr 01.
Artigo em Chinês | MEDLINE | ID: mdl-18756950

RESUMO

OBJECTIVE: To investigate whether erythropoietin (EPO) protects neuron against ketamine induced injuries. METHODS: Neurons were obtained from SD rat brain, cultured, and treated with ketamine of the concentrations of 0.1, 1, 10, and 30 micromol/L respectively. Neurons not treated by any agent were used as control group. Another neurons were divided into 3 groups undergoing the treatment of ketamine of the terminal concentration of 10 micromol/L, EPO + ketamine group undergoing the treatment of 10 micromol/L ketamine and EPO of the terminal concentrations of 0.3, 1, 3, and 10 U/ml, and ketamine + EPO + LY294002 group undergoing the treatment of 10 micromol/L ketamine, 1 U/ml EPO, and 10 micromol/L LY294002, a P13k inhibitor. Twenty-four hours after the co-inoculation the survival rates of the neurons were detected by MTT method. The apoptotic rate was detected by TUNEL assay. The neuron vitality was measured by MTT assay. Apoptotic neurons were measured by TUNEL assay. The activity of caspase-3 was detected with the caspase-3 fluorometric assay kit. The level of pAkt protein was analyzed by Western blotting. RESULTS: The survival rates of the neurons exposed to ketamine of the concentrations of 1, 10, and 30 micromol/L were (91 +/- 5)%, (42 +/- 6)%, and (23 +/- 7)% respectively, significantly lower than that of the control group (P < 0.05 or P < 0.01). The survival rates of the neurons treated by 10 micromol/L ketamine and EPO of the concentrations of 0.3, 1, 3, and 10 U/ml were (73 +/- 6)%, (86 +/- 9)%, (78 +/- 8)%, and (71 +/- 10)% respectively, all significantly high than that of the 10 micromol/L ketamine group (P < 0.05 or P < 0.01). The number of apoptotic neurons of the 10 micromol/L ketamine group was significantly higher than that pf the control group, the number of apoptotic neurons of the 10 micromol/L ketamine + 10 U/ml EPO group was significantly lower than that of the 10 micromol/L ketamine, and the number of apoptotic neurons of the ketamine + EPO + LY294002 group was (130 +/- 30)%, remarkably lower than that of the ketamine group. (P < 0.01), and the relative activity of caspase-3 of the 10 micromol/L ketamine group was (280 +/- 60)%, significantly higher than that of the control group, (P < 0.01). The relative activity of caspase-3 of the ketamine + EPO + LY294002 group was (220 +/- 34)%, significantly higher than that of the ketamine + EPO (P < 0.01). The pAkt protein level of the 10 micromol/L ketamine group was significantly lower than that of the control group (P < 0.05), the pAkt protein level of the 10 micromol/L ketamine + 10 U/ml EPO group was significantly higher than that of the 10 micromol/L ketamine group (P < 0.01), and the pAkt protein level of the ketamine + EPO + LY294002 group was significantly lower than that of the ketamine + 10 U/ml EPO group (P < 0.01). CONCLUSION: EPO affords significant neuroprotection against ketamine induced injury in neurons via PI3K/Akt-mediated signaling pathway.


Assuntos
Eritropoetina/farmacologia , Ketamina/toxicidade , Neurônios/efeitos dos fármacos , Animais , Apoptose , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Neurônios/citologia , Neurônios/metabolismo , Fosforilase b/biossíntese , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes
19.
Chin Med J (Engl) ; 131(10): 1214-1219, 2018 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-29722341

RESUMO

OBJECTIVE: Exposure to halogens, such as chlorine or bromine, results in environmental and occupational hazard to the lung and other organs. Chlorine is highly toxic by inhalation, leading to dyspnea, hypoxemia, airway obstruction, pneumonitis, pulmonary edema, and acute respiratory distress syndrome (ARDS). Although bromine is less reactive and oxidative than chlorine, inhalation also results in bronchospasm, airway hyperresponsiveness, ARDS, and even death. Both halogens have been shown to damage the systemic circulation and result in cardiac injury as well. There is no specific antidote for these injuries since the mechanisms are largely unknown. DATA SOURCES: This review was based on articles published in PubMed databases up to January, 2018, with the following keywords: "chlorine," "bromine," "lung injury," and "ARDS." STUDY SELECTION: The original articles and reviews including the topics were the primary references. RESULTS: Based on animal studies, it is found that inhaled chlorine will form chlorine-derived oxidative products that mediate postexposure toxicity; thus, potential treatments will target the oxidative stress and inflammation induced by chlorine. Antioxidants, cAMP-elevating agents, anti-inflammatory agents, nitric oxide-modulating agents, and high-molecular-weight hyaluronan have shown promising effects in treating acute chlorine injury. Elevated free heme level is involved in acute lung injury caused by bromine inhalation. Hemopexin, a heme-scavenging protein, when administered postexposure, decreases lung injury and improves survival. CONCLUSIONS: At present, there is an urgent need for additional research to develop specific therapies that target the basic mechanisms by which halogens damage the lungs and systemic organs.


Assuntos
Lesão Pulmonar Aguda/induzido quimicamente , Halogênios/toxicidade , Síndrome do Desconforto Respiratório/tratamento farmacológico , Animais , Cloro/toxicidade , Humanos , Pulmão/efeitos dos fármacos , Pulmão/patologia
20.
Chin Med J (Engl) ; 131(10): 1225-1231, 2018 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-29553050

RESUMO

OBJECTIVE: Mechanical ventilation (MV) has long been used as a life-sustaining approach for several decades. However, researchers realized that MV not only brings benefits to patients but also cause lung injury if used improperly, which is termed as ventilator-induced lung injury (VILI). This review aimed to discuss the pathogenesis of VILI and the underlying molecular mechanisms. DATA SOURCES: This review was based on articles in the PubMed database up to December 2017 using the following keywords: "ventilator-induced lung injury", "pathogenesis", "mechanism", and "biotrauma". STUDY SELECTION: Original articles and reviews pertaining to mechanisms of VILI were included and reviewed. RESULTS: The pathogenesis of VILI was defined gradually, from traditional pathological mechanisms (barotrauma, volutrauma, and atelectrauma) to biotrauma. High airway pressure and transpulmonary pressure or cyclic opening and collapse of alveoli were thought to be the mechanisms of barotraumas, volutrauma, and atelectrauma. In the past two decades, accumulating evidence have addressed the importance of biotrauma during VILI, the molecular mechanism underlying biotrauma included but not limited to proinflammatory cytokines release, reactive oxygen species production, complement activation as well as mechanotransduction. CONCLUSIONS: Barotrauma, volutrauma, atelectrauma, and biotrauma contribute to VILI, and the molecular mechanisms are being clarified gradually. More studies are warranted to figure out how to minimize lung injury induced by MV.


Assuntos
Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo , Animais , Barotrauma/metabolismo , Humanos , Espécies Reativas de Oxigênio/metabolismo , Ferimentos e Lesões/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa