Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
BMC Anesthesiol ; 24(1): 41, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38291353

RESUMO

BACKGROUND: Although previous studies have showed that epidural morphine can be used as a complement to local anesthetics for analgesia after postcesarean delivery under spinal anesthesia, there is little known about the analgesic dosage of epidural morphine and hydromorphone for hemorrhoidectomy. Therefore, we conducted this study to examine the potency ratio of hydromorphone to epidural morphine as well as effective analgesic dose for 50% patients (ED50) undergoing elective hemorrhoidectomy. METHODS: 80 patients under elective hemorrhoidectomy with combined spinal and epidural anesthesia(CSEA) in department of anesthesia, Dongguan Tungwah hospital. To assess the ED50, patients were treated with epidural morphine or epidural hydromorphone randomly using a biased coin method-determined dose with a sequential allocation procedure. Following surgery, standardized multimodal analgesia was administered to all patients. A pain response score of ≤ 3 (on a scale of 0-10) was determined to be the effective dose after 24 h following CSEA. The ED50 in both groups were determined using the probit regression and isotonic regression method. We also measured pain intensity by patient interview using a 10 point verbal numeric rating scale prospectively at 6, 12 and 24 h after CSEA, and adverse effects were also noted. RESULTS: The ED50 was 0.350 mg (95% CI, 0.259-0.376 mg) in hydromorphone group and 1.129 mg (95% CI, 0.903-1.187 mg) in morphine group, respectively, estimated by isotonic regression method. Regression analysis with the probit, the ED50 of epidural hydromorphone was 0.366 mg (95% CI, 0.276-0.388 mg) and epidural morphine was 1.138 mg (95% CI, 0.910-1.201 mg). Exploratory findings showed that there was no difference between the most frequent dosages of epidural hydromorphone or epidural morphine in the occurrence of nausea, vomiting and pruritus. When administered with epidural opioids at ED50 doses or higher, 97.5% (39/40) of epidural morphine patients and 97.5% (39/40) epidural hydromorphone of patients were satisfied with their analgesia. CONCLUSION: Effective hemorrhoidectomy analgesia requires a 3:1 ratio of epidural morphine to epidural hydromorphone. Both drugs provide excellent patient satisfaction.


Assuntos
Analgesia Epidural , Hemorroidectomia , Humanos , Hidromorfona , Morfina , Analgesia Epidural/métodos , Dor Pós-Operatória/epidemiologia , Analgésicos Opioides , Analgésicos/uso terapêutico , Método Duplo-Cego
2.
Anesth Analg ; 137(2): 426-439, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37145970

RESUMO

BACKGROUND: Myocardial infarction is a common perioperative complication, and blood flow restoration causes ischemia/reperfusion injury (IRI). Dexmedetomidine (DEX) pretreatment can protect against cardiac IRI, but the mechanism is still insufficiently understood. METHODS: In vivo, myocardial ischemia/reperfusion (30 minutes/120 minutes) was induced via ligation and then reperfusion of the left anterior descending coronary artery (LAD) in mice. Intravenous infusion of 10 µg/kg DEX was performed 20 minutes before ligation. Moreover, the α2-adrenoreceptor antagonist Yohimbine and STAT3 inhibitor Stattic were applied 30 minutes ahead of DEX infusion. In vitro, hypoxia/reoxygenation (H/R) with DEX pretreatment for 1 hour was performed in isolated neonatal rat cardiomyocytes. In addition, Stattic was applied before DEX pretreatment. RESULTS: In the mouse cardiac ischemia/reperfusion model, DEX pretreatment lowered the serum creatine kinase-MB isoenzyme (CK-MB) levels (2.47 ± 0.165 vs 1.55 ± 0.183; P < .0001), downregulated the inflammatory response ( P ≤ .0303), decreased 4-hydroxynonenal (4-HNE) production and cell apoptosis ( P = .0074), and promoted the phosphorylation of STAT3 (4.94 ± 0.690 vs 6.68 ± 0.710, P = .0001), which could be blunted by Yohimbine and Stattic. The bioinformatic analysis of differentially expressed mRNAs further confirmed that STAT3 signaling might be involved in the cardioprotection of DEX. Upon H/R treatment in isolated neonatal rat cardiomyocytes, 5 µM DEX pretreatment improved cell viability ( P = .0005), inhibited reactive oxygen species (ROS) production and calcium overload (both P ≤ .0040), decreased cell apoptosis ( P = .0470), and promoted STAT3 phosphorylation at Tyr705 (0.102 ± 0.0224 vs 0.297 ± 0.0937; P < .0001) and Ser727 (0.586 ± 0.177 vs 0.886 ± 0.0546; P = .0157), which could be abolished by Stattic. CONCLUSIONS: DEX pretreatment protects against myocardial IRI, presumably by promoting STAT3 phosphorylation via the α2-adrenoreceptor in vivo and in vitro.


Assuntos
Dexmedetomidina , Isquemia Miocárdica , Traumatismo por Reperfusão Miocárdica , Traumatismo por Reperfusão , Animais , Camundongos , Ratos , Apoptose , Creatina Quinase Forma MB , Dexmedetomidina/farmacologia , Modelos Animais de Doenças , Hipóxia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio , Transdução de Sinais , Receptores Adrenérgicos alfa
3.
Hypertens Res ; 46(8): 1934-1948, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37248323

RESUMO

Hypertension-induced renal injury is characterized by robust inflammation and tubulointerstitial fibrosis. Jumonji domain containing-3 (JMJD3) is closely linked with inflammatory response and fibrogenesis. Here we examined the effect of myeloid JMJD3 ablation on kidney inflammation and fibrosis in deoxycorticosterone acetate (DOCA)/salt hypertension. Our results showed that JMJD3 is notably induced in the kidneys with hypertensive injury. DOCA/salt stress causes an elevation in blood pressure that was no difference between myeloid specific JMJD3-deficient mice and wild-type control mice. Compared with wild-type control mice, myeloid JMJD3 ablation ameliorated kidney function and injury of mice in response to DOCA/salt challenge. Myeloid JMJD3 ablation attenuated collagen deposition, extracellular matrix proteins expression, and fibroblasts activation in injured kidneys following DOCA/salt treatment. Furthermore, myeloid JMJD3 ablation blunts inflammatory response in injured kidneys after DOCA/salt stress. Finally, myeloid JMJD3 ablation precluded myeloid myofibroblasts activation and protected against macrophages to myofibroblasts transition in injured kidneys. These beneficial effects were accompanied by reduced expression of interferon regulator factor 4. In summary, JMJD3 ablation in myeloid cells reduces kidney inflammation and fibrosis in DOCA salt-induced hypertension. Inhibition of myeloid JMJD3 may be a novel potential therapeutic target for hypertensive nephropathy. Myeloid JMJD3 deficiency reduces inflammatory response, myeloid fibroblasts activation, macrophages to myofibroblasts transition, and delays kidney fibrosis progression.


Assuntos
Acetato de Desoxicorticosterona , Hipertensão Renal , Hipertensão , Animais , Camundongos , Acetato de Desoxicorticosterona/efeitos adversos , Rim , Pressão Sanguínea , Inflamação/metabolismo , Macrófagos/metabolismo , Fibrose , Desoxicorticosterona/efeitos adversos , Desoxicorticosterona/metabolismo , Camundongos Endogâmicos C57BL
4.
BMC Anesthesiol ; 23(1): 105, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-37005591

RESUMO

BACKGROUND: The benefit of remote ischemia preconditioning (RIPreC) in pediatric cardiac surgery is unclear. The objective of this systematic review and meta-analysis was to examine the effectiveness of RIPreC in reducing the duration of mechanical ventilation and intensive care unit (ICU) length of stay after pediatric cardiac surgery. METHODS: We searched PubMed, EMBASE and the Cochrane Library from inception to December 31, 2022. Randomized controlled trials comparing RIPreC versus control in children undergoing cardiac surgery were included. The risk of bias of included studies was assessed using the Risk of Bias 2 (RoB 2) tool. The outcomes of interest were postoperative duration of mechanical ventilation and ICU length of stay. We conducted random-effects meta-analysis to calculate weighted mean difference (WMD) with 95% confidence interval (CI) for the outcomes of interest. We performed sensitivity analysis to examine the influence of intraoperative propofol use. RESULTS: Thirteen trials enrolling 1,352 children were included. Meta-analyses of all trials showed that RIPreC did not reduce postoperative duration of mechanical ventilation (WMD -5.35 h, 95% CI -12.12-1.42) but reduced postoperative ICU length of stay (WMD -11.48 h, 95% CI -20.96- -2.01). When only trials using propofol-free anesthesia were included, both mechanical ventilation duration (WMD -2.16 h, 95% CI -3.87- -0.45) and ICU length of stay (WMD -7.41 h, 95% CI -14.77- -0.05) were reduced by RIPreC. The overall quality of evidence was moderate to low. CONCLUSIONS: The effects of RIPreC on clinical outcomes after pediatric cardiac surgery were inconsistent, but both postoperative mechanical ventilation duration and ICU length of stay were reduced in the subgroup of children not exposed to propofol. These results suggested a possible interaction effect of propofol. More studies with adequate sample size and without intraoperative propofol use are needed to define the role of RIPreC in pediatric cardiac surgery.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Precondicionamento Isquêmico , Propofol , Criança , Humanos , Unidades de Terapia Intensiva , Tempo de Internação , Respiração Artificial
5.
J Hypertens ; 41(5): 794-810, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36883469

RESUMO

BACKGROUND: Inflammation and renal interstitial fibrosis are the main pathological features of hypertensive nephropathy. Interferon regulatory factor 4 (IRF-4) has an important role in the pathogenesis of inflammatory and fibrotic diseases. However, its role in hypertension-induced renal inflammation and fibrosis remains unexplored. METHOD AND RESULTS: We showed that deoxycorticosterone acetate (DOCA)-salt resulted in an elevation of blood pressure and that there was no difference between wild-type and IRF-4 knockout mice. IRF-4 -/- mice presented less severe renal dysfunction, albuminuria, and fibrotic response after DOCA-salt stress compared with wild-type mice. Loss of IRF-4 inhibited extracellular matrix protein deposition and suppressed fibroblasts activation in the kidneys of mice subjected to DOCA-salt treatment. IRF-4 disruption impaired bone marrow-derived fibroblasts activation and macrophages to myofibroblasts transition in the kidneys in response to DOCA-salt treatment. IRF-4 deletion impeded the infiltration of inflammatory cells and decreased the production of proinflammatory molecules in injured kidneys. IRF-4 deficiency activated phosphatase and tensin homolog and weakened phosphoinositide-3 kinase/AKT signaling pathway in vivo or in vitro . In cultured monocytes, TGFß1 also induced expression of fibronectin and α-smooth muscle actin and stimulated the transition of macrophages to myofibroblasts, which was blocked in the absence of IRF-4. Finally, macrophages depletion blunted macrophages to myofibroblasts transition, inhibited myofibroblasts accumulation, and ameliorated kidney injury and fibrosis. CONCLUSION: Collectively, IRF-4 plays a critical role in the pathogenesis of kidney inflammation and fibrosis in DOCA-salt hypertension.


Assuntos
Acetato de Desoxicorticosterona , Hipertensão Renal , Hipertensão , Animais , Camundongos , Acetatos/efeitos adversos , Acetatos/metabolismo , Pressão Sanguínea , Desoxicorticosterona/efeitos adversos , Desoxicorticosterona/metabolismo , Acetato de Desoxicorticosterona/efeitos adversos , Fibrose , Hipertensão/etiologia , Hipertensão Renal/metabolismo , Inflamação/metabolismo , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Rim , Camundongos Knockout
6.
Shock ; 58(6): 556-564, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36374735

RESUMO

ABSTRACT: Background: Dexmedetomidine (DEX) attenuates intestinal I/R injury, but its mechanism of action remains to be further elucidated. Protein disulfide isomerase A3 (PDIA3) has been reported as a therapeutic protein for the prevention and treatment of intestinal I/R injury. This study was to investigate whether PDIA3 is involved in intestinal protection of DEX and explore the underlying mechanisms. Methods: The potential involvement of PDIA3 in DEX attenuation of intestinal I/R injury was tested in PDIA3 Flox/Flox mice and PDIA3 conditional knockout (cKO) in intestinal epithelium mice subjected to 45 min of superior mesenteric artery occlusion followed by 4 h of reperfusion. Furthermore, the α2-adrenergic receptor (α2-AR) antagonist, yohimbine, was administered in wild-type C57BL/6N mice intestinal I/R model to investigate the role of α2-AR in the intestinal protection conferred by DEX. Results: In the present study, we identified intestinal I/R-induced obvious inflammation, endoplasmic reticulum (ER) stress-dependent apoptosis, and oxidative stress, and all the aforementioned changes were improved by the administration of DEX. PDIA3 cKO in the intestinal epithelium have reversed the protective effects of DEX. Moreover, yohimbine also reversed the intestinal protection of DEX and downregulated the messenger RNA and protein levels of PDIA3. Conclusion: DEX prevents PDIA3 decrease by activating α2-AR to inhibit intestinal I/R-induced inflammation, ER stress-dependent apoptosis, and oxidative stress in mice.


Assuntos
Dexmedetomidina , Animais , Camundongos , Dexmedetomidina/farmacologia , Dexmedetomidina/uso terapêutico , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos alfa 2/metabolismo , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/farmacologia , Camundongos Endogâmicos C57BL , Apoptose , Ioimbina/farmacologia , Inflamação/tratamento farmacológico
7.
Front Pharmacol ; 13: 1019109, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36278213

RESUMO

Gut-vascular barrier (GVB) serves as the last barrier to limit the migration of intestinal toxins into the blood circulation. The efficacy of terlipressin (a vasopressin V1 receptor agonist) in reducing GVB and multiple organ damage in gut-derived sepsis is unknown. In this study, we hypothesized that, besides other intestinal barriers, GVB play a key role in gut-derived sepsis and terlipressin improve GVB damage and then reduce bacterial translocation and organ injuries. In vivo, a cecal ligation and puncture mouse model was established. The mice were subjected to examine the damage of GVB determined by intestinal plasmalemma vesicle-associated protein-1(PV-1) and vascular endothelial-cadherin. And the intestinal permeability was assessed by translocation of intestinal bacteria and macromolecules. In vitro, transendothelial electrical resistance (TER) during interleukin (IL)-1ß stimulation was measured on endothelial cells with or without small interfering RNA targeting ß-catenin (si ß-catenin). Terlipressin significantly improved GVB damage and reduced translocation of intestinal macromolecules and bacteria by activating PI3K signaling. Of note, intestinal PV-1 expression was significantly correlated with translocation of macromolecules, and dramatic increase of macromolecules was observed in intestinal tissues whereas fewer macromolecules and bacteria were observed in blood, liver and lung following terlipressin treatment. In vitro, terlipressin restored TER during IL-1ß stimulation and si ß-catenin transfection blocked the changes delivered by terlipressin. Collectively, terlipressin alleviated GVB damage and subsequent bacterial translocation via blood vessels after sepsis challenge, resulting in reduced distant organ injuries and the responsible mechanisms may involve the activation of PI3K/ß-catenin pathway.

8.
Front Immunol ; 13: 978262, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36159833

RESUMO

Renal fibrosis commonly occurs in the process of chronic kidney diseases. Here, we explored the role of Jumonji domain containing 3 (Jmjd3)/interferon regulatory factor 4 (IRF4) axis in activation of myeloid fibroblasts and transition of M2 macrophages into myofibroblasts transition (M2MMT) in kidney fibrosis. In mice, Jmjd3 and IRF4 were highly induced in interstitial cells of kidneys with folic acid or obstructive injury. Jmjd3 deletion in myeloid cells or Jmjd3 inhibitor reduced the levels of IRF4 in injured kidneys. Myeloid Jmjd3 depletion impaired bone marrow-derived fibroblasts activation and M2MMT in folic acid or obstructive nephropathy, resulting in reduction of extracellular matrix (ECM) proteins expression, myofibroblasts formation and renal fibrosis progression. Pharmacological inhibition of Jmjd3 also prevented myeloid fibroblasts activation, M2MMT, and kidney fibrosis development in folic acid nephropathy. Furthermore, IRF4 disruption inhibited myeloid myofibroblasts accumulation, M2MMT, ECM proteins accumulation, and showed milder fibrotic response in obstructed kidneys. Bone marrow transplantation experiment showed that wild-type mice received IRF4-/- bone marrow cells presented less myeloid fibroblasts activation in injured kidneys and exhibited much less kidney fibrosis after unilateral ureteral obstruction. Myeloid Jmjd3 deletion or Jmjd3 inhibitor attenuated expressions of IRF4, α-smooth muscle actin and fibronectin and impeded M2MMT in cultured monocytes exposed to IL-4. Conversely, overexpression IRF4 abrogated the effect of myeloid Jmjd3 deletion on M2MMT. Thus, Jmjd3/IRF4 signaling has a crucial role in myeloid fibroblasts activation, M2 macrophages to myofibroblasts transition, extracellular matrix protein deposition, and kidney fibrosis progression.


Assuntos
Miofibroblastos , Insuficiência Renal Crônica , Actinas/metabolismo , Animais , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Fibrose , Ácido Fólico/farmacologia , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Interleucina-4/metabolismo , Histona Desmetilases com o Domínio Jumonji , Macrófagos/metabolismo , Camundongos , Miofibroblastos/metabolismo , Insuficiência Renal Crônica/patologia
9.
Front Pharmacol ; 13: 940716, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35924048

RESUMO

Renal fibrosis is an important pathological biomarker of chronic kidney disease (CKD). Stimulator of interferon genes/TANK binding kinase 1 (STING/TBK1) axis has been identified as the main regulator of innate immune response and closely related to fibrotic disorder. However, the role of STING/TBK1 signaling pathway in kidney fibrosis is still unknown. In this study, we investigated the effect of pharmacological inhibition of STING/TBK1 signaling on renal fibrosis induced by folic acid (FA). In mice, TBK1 was significantly activated in interstitial cells of FA-injured kidneys, which was markedly inhibited by H-151 (a STING inhibitor) treatment. Specifically, pharmacological inhibition of STING impaired bone marrow-derived fibroblasts activation and macrophage to myofibroblast transition in folic acid nephropathy, leading to reduction of extracellular matrix proteins expression, myofibroblasts formation and development of renal fibrosis. Furthermore, pharmacological inhibition of TBK1 by GSK8612 reduced myeloid myofibroblasts accumulation and impeded macrophage to myofibroblast differentiation, resulting in less deposition of extracellular matrix protein and less severe fibrotic lesion in FA-injured kidneys. In cultured mouse bone marrow-derived monocytes, TGF-ß1 activated STING/TBK1 signaling. This was abolished by STING or TBK1 inhibitor administration. In addition, GSK8612 treatment decreased levels of α-smooth muscle actin and extracellular matrix proteins and prevents bone marrow-derived macrophages to myofibroblasts transition in vitro. Collectively, our results revealed that STING/TBK1 signaling has a critical role in bone marrow-derived fibroblast activation, macrophages to myofibroblasts transition, and kidney fibrosis progression.

10.
Front Cell Dev Biol ; 10: 818513, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35155431

RESUMO

Background: Remifentanil protects against intestinal ischemia/reperfusion (I/R) injury; however, its exact mechanism remains to be elucidated. The objective of this study was to investigate the underlying molecular mechanism of remifentanil in intestinal I/R injury in mice. Methods: We evaluated the intestine-protective effect of remifentanil in adult male mice with 45 min superior mesenteric artery occlusion followed by 4 h reperfusion by determining the following: intestinal Chiu's scores, diamine oxidase, and intestinal fatty acid binding protein in serum; the apoptotic index, lipid peroxidation product malondialdehyde (MDA), and superoxide dismutase (SOD) activity in the intestinal mucosa; and the intestinal mRNA and protein expressions of Bip, CHOP, caspase-12, and cleaved caspase-3, reflecting endoplasmic reticulum (ER) stress. Furthermore, conditional knockout mice, in which the protein disulfide isomerase A3 (PDIA3) gene was deleted from the intestinal epithelium, and SB203580 (a selective p38MAPK inhibitor) were used to determine the role of PDIA3 and p38MAPK in I/R progression and intestinal protection by remifentanil. Results: Our data showed that intestinal I/R induced obvious oxidative stress and endoplasmic reticulum stress-related cell apoptosis, as evidenced by an increase in the intestinal mucosal malondialdehyde, a decrease in the intestinal mucosal SOD, and an increase in the apoptotic index and the mRNA and protein expression of Bip, CHOP, caspase-12, and cleaved caspase-3. Remifentanil significantly improved these changes. Moreover, the deletion of intestinal epithelium PDIA3 blocked the protective effects of remifentanil. SB203580 also abolished the intestinal protection of remifentanil and downregulated the mRNA and protein expression of PDIA3. Conclusion: Remifentanil appears to act via p38MAPK to protect the small intestine from intestinal I/R injury by its PDIA3-mediated antioxidant and anti-ER stress properties.

11.
J Immunol ; 208(4): 968-978, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35063996

RESUMO

Influx of activated neutrophils into the lungs is the histopathologic hallmark of acute lung injury (ALI) after intestinal ischemia/reperfusion (I/R). Neutrophils can release DNA and granular proteins to form cytotoxic neutrophil extracellular traps (NETs), which promotes bystander tissue injury. However, whether NETs are responsible for the remote ALI after intestinal I/R and the mechanisms underlying the dissemination of harmful gut-derived mediators to the lungs are unknown. In the C57BL/6J mouse intestinal I/R model, DNase I-mediated degradation and protein arginine deiminase 4 (PAD4) inhibitor-mediated inhibition of NET treatments reduced NET formation, tissue inflammation, and pathological injury in the lung. High-mobility group protein B1 (HMGB1) blocking prevented NET formation and protected against tissue inflammation, as well as reduced cell apoptosis and improved survival rate. Moreover, recombinant human HMGB1 administration further drives NETs and concurrent tissue toxic injury, which in turn can be reversed by neutrophil deletion via anti-Ly6G Ab i.p. injection. Furthermore, global MyD88 deficiency regulated NET formation and alleviated the development of ALI induced by intestinal I/R. Thus, HMGB1 released from necroptotic enterocytes caused ALI after intestinal I/R by inducing NET formation. Targeting NETosis and the HMGB1 pathway might extend effective therapeutic strategies to minimize intestinal I/R-induced ALI.


Assuntos
Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/metabolismo , Armadilhas Extracelulares/genética , Proteína HMGB1/genética , Neutrófilos/imunologia , Neutrófilos/metabolismo , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Apoptose/genética , Biomarcadores , Modelos Animais de Doenças , Suscetibilidade a Doenças , Proteína HMGB1/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Traumatismo por Reperfusão/patologia
12.
Anesth Analg ; 134(2): 419-431, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34889823

RESUMO

BACKGROUND: Intestinal ischemia/reperfusion (I/R) challenge often results in gut barrier dysfunction and induces distant organ injury. Dexmedetomidine has been shown to protect intestinal epithelial barrier against I/R attack. The present study aims to investigate the degree to which intestinal I/R attack will contribute to gut-vascular barrier (GVB) damage, and to examine the ability of dexmedetomidine to minimize GVB and liver injuries in mice. METHODS: In vivo, intestinal ischemic challenge was induced in mice by clamping the superior mesenteric artery for 45 minutes. After clamping, the mice were subjected to reperfusion for either 2, 4, 6, or 12 hours. Intraperitoneal injection of dexmedetomidine 15, 20, or 25 µg·kg-1 was performed intermittently at the phase of reperfusion. For the in vitro experiments, the challenge of oxygen-glucose deprivation/reoxygenation (OGD/R) was established in cultured vascular endothelial cells, and dexmedetomidine (1 nM) was used to treat the cells for 24 hours. Moreover, in vivo and in vitro, SKL2001 (a specific agonist of ß-catenin) or XAV939 (a specific inhibitor of ß-catenin) was applied to determine the role of ß-catenin in the impacts provided by dexmedetomidine. RESULTS: The attack of intestinal I/R induced GVB damage. The greatest level of damage was observed at 4 hours after intestinal reperfusion. There was a significant increase in plasmalemma vesicle-associated protein-1 (PV1, a specific biomarker for endothelial permeability) expression (5.477 ± 0.718 vs 1.000 ± 0.149; P < .001), and increased translocation of intestinal macromolecules and bacteria to blood and liver tissues was detected (all P < .001). Liver damages were observed. There were significant increases in histopathological scores, serum parameters, and inflammatory factors (all P < .001). Dexmedetomidine 20 µg·kg-1 reduced PV1 expression (0.466 ± 0.072 vs 1.000 ± 0.098; P < .001) and subsequent liver damages (all P < .01). In vitro, dexmedetomidine significantly improved vascular endothelial cell survival (79.387 ± 6.447% vs 50.535 ± 1.766%; P < .001) and increased the productions of tight junction protein and adherent junction protein (all P < .01) following OGD/R. Importantly, in cultured cells and in mice, ß-catenin expression significantly decreased (both P < .001) following challenge. Dexmedetomidine or SKL2001 upregulated ß-catenin expression and produced protective effects (all P < .01). However, XAV939 completely eliminated the protective effects of dexmedetomidine on GVB (all P < .001). CONCLUSIONS: The disruption of GVB occurred following intestinal I/R. Dexmedetomidine alleviated I/R-induced GVB impairment and subsequent liver damage.


Assuntos
Analgésicos não Narcóticos/administração & dosagem , Permeabilidade Capilar/efeitos dos fármacos , Dexmedetomidina/administração & dosagem , Mucosa Intestinal/efeitos dos fármacos , Hepatopatias/tratamento farmacológico , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Permeabilidade Capilar/fisiologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Injeções Intraperitoneais , Mucosa Intestinal/metabolismo , Hepatopatias/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão/metabolismo
13.
Ren Fail ; 43(1): 1368-1377, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34602010

RESUMO

INTRODUCTION: Advancing renal fibrosis is the common histopathological feature of chronic obstructive nephropathy, representing the final pathway of nearly all chronic and progressive nephropathies. Increasing evidences suggest that circular RNAs (circRNAs) are crucial regulatory molecules present at virtually every level of the cellular pathophysiological process. Nonetheless, there are a few evidences for the role of circRNAs in renal fibrosis induced by obstructive nephropathy. AIMS: We performed RNA-seq analysis to analyze the expression profiles of circRNAs in the obstructed kidneys to identify the potential circRNAs and their network. METHODS: With silk ligated the left ureter to establish a mice unilateral ureteral obstruction (UUO) model. Renal tissue circRNAs were obtained and were screened by a circRNA microarray. The circRNA-miRNA-mRNA regulatory network and the target genes were visualized using Cytoscape software. RESULTS: The microarray results showed that 5454 and 2935 circRNAs were detected in the control and UUO group, respectively. There were 605 circRNAs up-regulated and 745 circRNAs down-regulated in the obstructive kidneys. The top 5 up-regulated and down-regulated circRNAs were chosen for predicting the circRNA/miRNA/target mRNAs triple network. The GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis showed that these circRNAs and the triple network were enriched in the process of apoptosis, p53 signaling pathway, cell growth and cell death, which might participate in the pathogenesis of obstructive nephrology. CONCLUSION: Our results show that the dis-regulated circRNAs might play crucial roles in the pathogenesis of obstructive nephropathy, which proceeds to identify novel therapeutic targets for chronic kidney disease.


Assuntos
Rim/patologia , MicroRNAs/genética , RNA Circular/genética , RNA Mensageiro/genética , Obstrução Ureteral/genética , Animais , Apoptose/genética , Biologia Computacional/métodos , Modelos Animais de Doenças , Fibrose/patologia , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Obstrução Ureteral/patologia
14.
J Surg Res ; 267: 687-694, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34274907

RESUMO

BACKGROUND: Pneumoperitoneum during laparoscopic surgery has a systemic impact on the renal system and might contribute to acute kidney injury or postoperative renal dysfunction. However, effective preventive strategies are still lacking. We aimed to explore the effects of dexmedetomidine (DEX) on kidney and other organ function in patients undergoing elective laparoscopic surgery for colorectal cancer. MATERIALS AND METHODS: Fifty-six patients were randomly enrolled into the Control or DEX group. The DEX group received 1 µg kg-1 DEX intravenously within 10 min followed by a maintenance dose of 0.5 µg kg-1 h-1 infused until 30 min before closing the peritoneum. In the Control group, 0.9% sodium chloride was administered as a placebo. The primary outcome was serum neutrophil gelatinase-associated lipocalin (NGAL) levels reflecting kidney injury. Secondary outcomes included variables reflecting the kidney, intestinal injury and systemic inflammatory response. RESULTS: NGAL levels were significantly lower in the DEX group than in the Control group at 1 d and 5 d postoperatively (107.5 ± 55.6 ng mL-1versus 179.5 ± 78.2 ng mL-1; 70.3 ± 45.8 ng mL-1versus 135.2 ± 59.6 ng mL-1, P < 0.001), while the BUN and Cr levels showed no differences between the groups. Serum DAO activity was significantly lower in the DEX group patients 24 h after surgery. Moreover, I-FABP levels were markedly lower at 2 h and 24 h postoperatively in the DEX group than in the Control group (P < 0.001). CONCLUSIONS: Perioperative DEX administration may potentially confer kidney and intestinal protection during laparoscopic surgery for colorectal cancer patients.


Assuntos
Injúria Renal Aguda , Neoplasias Colorretais , Dexmedetomidina , Laparoscopia , Injúria Renal Aguda/prevenção & controle , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/cirurgia , Dexmedetomidina/farmacologia , Humanos , Rim , Laparoscopia/efeitos adversos
15.
Int Immunopharmacol ; 98: 107907, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34243040

RESUMO

Renal fibrosis is a histological manifestation of chronic kidney disease. Natural killer T (NKT) cells have a critical role in the pathogenesis of fibrotic disorder. However, the role of NKT cells in regulating kidney fibrosis remains largely unknown. In the current study, we showed that the percentages of NKT+ cells and NKT+-IL-4+ cells were notably increased in folic acid (FA) and obstructive nephropathy. CD1d deficiency protected mice from renal fibrosis induced by FA and obstructive injury. Specifically, Loss of CD1d reduced bone marrow-derived myofibroblasts and CD206+/α-smooth muscle actin+ cells in the kidneys of injured mice. But mice treated with α-galactosylceramide (α-GC, a specific activator of NKT cells) developed more severe fibrosis, accumulated more myeloid myofibroblasts and M2 macrophages-myofibroblasts transition (M2MMT) cells in FA injured kidneys. Furthermore, IL-4 expression was markedly reduced in CD1d deficiency mice but increased in α-GC-treated mice. Administration of IL-4 abrogates the inhibiting effect of CD1d deficiency on renal fibrosis, bone marrow-derived fibroblasts activation, and M2MMT in FA injured kidneys. Conversely, pharmacological inhibition of IL-4 attenuated the development of renal fibrosis, decreased bone marrow-derived myofibroblasts, and suppressed M2MMT. Thus, this study revealed a novel role of NKT cells in the bone marrow-derived fibroblasts activation and M2MMT during renal fibrosis. Targeting NKT cell/IL-4 signaling may be an effective treatment for renal fibrosis.


Assuntos
Interleucina-4/metabolismo , Rim/patologia , Células T Matadoras Naturais/imunologia , Insuficiência Renal Crônica/imunologia , Animais , Antígenos CD1d/genética , Comunicação Celular/imunologia , Modelos Animais de Doenças , Fibrose , Ácido Fólico/administração & dosagem , Ácido Fólico/toxicidade , Humanos , Rim/efeitos dos fármacos , Rim/imunologia , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Miofibroblastos/imunologia , Miofibroblastos/patologia , Células T Matadoras Naturais/metabolismo , Insuficiência Renal Crônica/induzido quimicamente , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia
17.
Eur J Pharmacol ; 901: 174097, 2021 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-33848540

RESUMO

Renal fibrosis is the common pathological hallmark of chronic kidney disease, and SET domain containing lysine methyltransferase 7 (SETD7) promote considerably renal fibrosis. However, the signaling mechanisms underlying SETD7 driving renal fibrosis are not fully understood. Here, we investigated the role of SETD7 in M2 macrophages-myofibroblasts transition and the myeloid fibroblasts activation in folic acid and obstruction-induced renal fibrosis. Mice treated with PFI-2, an inhibitor of SETD7, presented less bone marrow-derived myofibroblasts, fewer CD206+/α-smooth muscle actin + cells and developed less renal fibrosis (P<0.01). Furthermore, SETD7 inhibition reduced the infiltration of inflammatory cells and decreased the production of pro-inflammatory cytokines and chemokines in the kidneys after folic acid treatment (P<0.01). Finally, SETD7 inhibition suppressed the accumulation of NF-κB p65+ cells in folic acid nephropathy (P<0.01). Taken together, SETD7 mediates M2 macrophages-myofibroblasts transition, bone marrow-derived myofibroblasts activation, and inflammation response in the development of renal fibrosis.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Ácido Fólico/farmacologia , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Isoquinolinas/farmacologia , Nefropatias/tratamento farmacológico , Rim/patologia , Sulfonamidas/farmacologia , Animais , Fibroblastos/efeitos dos fármacos , Fibrose , Nefropatias/induzido quimicamente , Nefropatias/patologia , Testes de Função Renal , Lectinas Tipo C/metabolismo , Macrófagos/efeitos dos fármacos , Masculino , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Superfície Celular/metabolismo , Fator de Transcrição RelA/efeitos dos fármacos
18.
Surgery ; 170(1): 239-248, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33745733

RESUMO

BACKGROUND: Ischemia/reperfusion of the intestine often leads to distant organ injury, but the mechanism of intestinal ischemia/reperfusion-induced renal dysfunction is still not clear. The present study aimed to investigate the mechanisms of acute renal damage after intestinal ischemia/reperfusion challenge and explore the role of released high-mobility group box-1 in this process. METHODS: Intestinal ischemia/reperfusion was induced in male Sprague-Dawley rats by clamping the superior mesenteric artery for 1.5 hours. At different reperfusion time points, anti-high-mobility group box-1 neutralizing antibodies or ethyl pyruvate were administered to neutralize or inhibit circulating high-mobility group box-1, respectively. RESULTS: Significant kidney injury was observed after 6 hours of intestinal reperfusion, as indicated by increased serum levels of urea nitrogen and creatinine, increased expression of neutrophil gelatinase-associated lipocalin, interleukin-6, and MIP-2, and enhanced cell apoptosis, as indicated by cleaved caspase 3 levels in renal tissues. The levels of phosphorylated eIF2ɑ, activating transcription factor 4, and C/EBP-homologous protein (CHOP) were markedly elevated, indicating the activation of endoplasmic reticulum stress in the impaired kidney. High-mobility group box-1 translocated to cytoplasm in the intestine and serum concentrations of high-mobility group box-1 increased notably during the reperfusion phase. Both anti-high-mobility group box-1 antibodies and ethyl pyruvate treatment significantly reduced serum high-mobility group box-1 concentrations, attenuated endoplasmic reticulum stress in renal tissue and inhibited the development of renal damage. Moreover, the elevated expression of receptor for advanced glycation end products in the kidneys after intestinal ischemia/reperfusion was abrogated after high-mobility group box-1 inhibition. CONCLUSION: These results suggested that high-mobility group box-1 signaling regulated endoplasmic reticulum stress and promoted intestinal ischemia/reperfusion-induced acute kidney injury. High-mobility group box-1 neutralization/inhibition might serve as a pharmacological intervention strategy for these pathophysiological processes.


Assuntos
Injúria Renal Aguda/etiologia , Estresse do Retículo Endoplasmático/fisiologia , Proteína HMGB1/metabolismo , Intestinos/patologia , Traumatismo por Reperfusão/complicações , Animais , Apoptose , Creatinina/sangue , Modelos Animais de Doenças , Intestinos/irrigação sanguínea , Isquemia/metabolismo , Rim/metabolismo , Masculino , Ratos Sprague-Dawley , Reperfusão/efeitos adversos , Transdução de Sinais , Fator de Transcrição CHOP/metabolismo
19.
ACS Biomater Sci Eng ; 6(10): 5857-5865, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-33320563

RESUMO

Identifying severe acute pancreatitis (SAP) as soon as possible is critical for achieving optimal outcomes and saving lives. In this study, a novel P-selectin-targeted, NIR fluorescent dye (Cy 5.5)-labeled dual-modal nanoprobe based on diethylenetriaminepentaacetic chelates (Gd-DTPA-Cy5.5-PsLmAb) was constructed for the bimodal imaging of SAP at the early stage. Gd-DTPA-Cy5.5-PsLmAb was prepared, and its structure was characterized by Fourier transform infrared spectroscopy, UV-vis spectroscopy, and fluorescence spectroscopy, and its stability was evaluated. Biocompatibility was evaluated by the hemolysis and cytotoxicity assays. The enzyme-linked immunosorbent assay was used to detect and evaluate the expression of P-selectin in the peripheral blood of 11 patients with acute pancreatitis (AP) and 5 healthy volunteers. The bimodal imaging ability of Gd-DTPA-Cy5.5-PsLmAb nanoprobes was evaluated via near-infrared fluorescence (NIRF) and magnetic resonance imaging (MRI) in AP animal models in vivo. Gd-DTPA-Cy5.5-PsLmAb showed low toxicity to human embryonic kidney cells (293T cells) and good blood compatibility. The P-selectin levels of humans and rats in the mild acute pancreatitis (MAP)/SAP stage were significantly higher than those in the control group and reached the highest level at the SAP stage. Furthermore, Gd-DTPA-Cy5.5-PsLmAb nanoprobes showed clear NIRF imaging of mouse pancreas at the MAP stage and SAP stage by a fluorescence signal at 6.09 × 108 and 1.95 × 109, respectively. Meanwhile, Gd-DTPA-Cy5.5-PsLmAb nanoprobes also successfully showed the T1-weighted MR signal of rat pancreas at the MAP stage, but Gd-DTPA seldom showed any signal increase at the MAP stage; Gd-DTPA-Cy5.5-PsLmAb and Gd-DTPA could show an increasing MR signal of rat pancreas at the SAP stage. Gd-DTPA-Cy5.5-PsLmAb proved to offer a stronger signal than Gd-DTPA.Our findings indicate that Gd-DTPA-Cy5.5-PsLmAb is an effective and specific MR/NIRF dual nanoprobe for bimodal imaging, providing a promising diagnostic approach for early SAP in clinic.


Assuntos
Selectina-P , Pancreatite , Doença Aguda , Animais , Gadolínio DTPA , Humanos , Imageamento por Ressonância Magnética , Pancreatite/induzido quimicamente , Ratos
20.
Int J Med Sci ; 17(17): 2751-2762, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33162803

RESUMO

Background: To date, the effect of vasopressin on organ damages after acute mesenteric ischemia (MI) remains poorly understood. Aims: To investigate the effect of terlipressin, a selective vasopressin V1 receptor agonist, versus norepinephrine on the intestinal and renal injuries after acute MI, and to explore the underlying mechanism of terlipressin. Methods: Acute MI model was produced by clamping the superior mesenteric artery for 1 hour. Immediately after unclamping, terlipressin or norepinephrine was intravenously administered for 2 hours. Meanwhile, in vitro, RAW264.7 cells were treated with lipopolysaccharide or lipopolysaccharide+terlipressin. In addition, wortmannin was used to determine the role of phosphoinositide 3-kinase (PI3K)/ protein kinase B (Akt) pathway in the potential impacts of terlipressin. Results: MI led to severe hypotension, caused notable intestinal and renal impairments and resulted in high mortality, which were markedly improved by terlipressin or norepinephrine. Terlipressin increased mean arterial pressure, decreased intestinal epithelial cell apoptosis, inhibited the generation of M1 macrophage in intestinal and renal tissues, and hindered the release of inflammatory cytokines after MI. Moreover, in cultured macrophages, terlipressin reduced the mRNA level of specific M1 markers and the release of inflammatory cytokines caused by lipopolysaccharide challenge. Wortmannin decreased the expression of PI3K and Akt induced by terlipressin in cells and in tissues, and abolished the above protective effects conferred by terlipressin. Conclusions: Terlipressin or norepinephrine could effectively improve organ damages and mortality after acute MI. Terlipressin elevates blood pressure and inhibits intestinal epithelial apoptosis and macrophage M1 polarization via the PI3K/Akt pathway.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Isquemia Mesentérica/tratamento farmacológico , Receptores de Vasopressinas/agonistas , Traumatismo por Reperfusão/tratamento farmacológico , Terlipressina/administração & dosagem , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/patologia , Animais , Apoptose/efeitos dos fármacos , Pressão Arterial/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Íleo/irrigação sanguínea , Íleo/efeitos dos fármacos , Íleo/patologia , Mucosa Intestinal/irrigação sanguínea , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Rim/irrigação sanguínea , Rim/efeitos dos fármacos , Rim/patologia , Masculino , Isquemia Mesentérica/complicações , Isquemia Mesentérica/patologia , Norepinefrina/administração & dosagem , Fosfatidilinositol 3-Quinase/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/patologia , Organismos Livres de Patógenos Específicos , Wortmanina/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...