Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
BMC Anesthesiol ; 22(1): 382, 2022 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-36482299

RESUMO

BACKGROUND: The mortality rate is high in critically ill patients due to the difficulty of diagnosis and treatment. Thus, it is very important to explore the predictive value of different indicators related to prognosis in critically ill patients. METHODS: This was a retrospective cohort study of patients in the intensive care unit (ICU) of the Sixth People's Hospital in Shanghai, China. A total of 1465 ICU patients had lactate values > 2.1 mmol/L at least once within 24 h of ICU admission, and arterial blood gas was monitored more than twice during the ICU stay. RESULTS: The predictive value of lactate clearance at 24 h was not high, and the sensitivity and specificity were lower. The predictive value of the lactate level at baseline and the APACHE II score was higher than that of lactate clearance at 24 h in critically ill patients. The predictive value of the lactate level at baseline combined with the APACHE II score was higher than that of the lactate level at baseline or the APACHE II score alone. In addition, the predictive value of lactate clearance at 24 h combined with the APACHE II score was also significantly higher than that of lactate clearance at 24 h or the APACHE II score alone. In particular, the area under the ROC curve reached 0.900, the predictive value was markedly higher than that of the ROC alone, and the sensitivity and specificity were better when these three indicators were combined. CONCLUSIONS: The combination of lactate level, lactate clearance and APACHE II score better predicts short-term outcomes in critically ill patients.


Assuntos
Ácido Láctico , Humanos , APACHE , Estudos Retrospectivos , China
2.
BMC Health Serv Res ; 20(1): 300, 2020 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-32293425

RESUMO

BACKGROUND: A reasonable allocation of health resources is often characterized by equity and high efficiency. This study aims to evaluate the equity and efficiency of maternal and child health (MCH) resources allocation in Hunan Province, China. METHODS: Data related to MCH resources and services was obtained from the Hunan maternal and child health information reporting and management system. The Gini coefficient and data envelopment analysis (DEA) were employed to evaluate the equity and efficiency of MCH resources allocation, respectively. RESULTS: The MCH resources allocation in terms of demographic dimension were in a preferred equity status with the Gini values all less than 0.3, and the Gini values for each MCH resources' allocation in terms of the geographical dimension ranged from 0.1298 to 0.4256, with the highest values in the number of midwives and medical equipment (≥ CNY 10,000), which exceeds 0.4, indicating an alert of inequity. More than 40% regions in Hunan were found to be relatively inefficient with decreased return to scale in the allocation of MCH resources, indicating those inefficient regions were using more inputs than needed to obtain the current output levels. CONCLUSIONS: The equity of MCH resources by population size is superior by geographic area and the disproportionate distribution of the number of medical equipment (≥ CNY 10,000) and midwives between different regions was the main source of inequity. Policy-makers need to consider the geographical accessibility of health resources among different regions to ensure people in different regions could get access to available health services. More than 40% of regions in Hunan were found to be inefficient, with using more health resources than needed to produce the current amount of health services. Further investigations on factors affecting the efficiency of MCH resources allocation is still needed to guide regional health plans-making and resource allocation.


Assuntos
Eficiência Organizacional , Alocação de Recursos para a Atenção à Saúde/organização & administração , Equidade em Saúde , Serviços de Saúde Materno-Infantil/organização & administração , Criança , China , Feminino , Acessibilidade aos Serviços de Saúde , Humanos , Gravidez
3.
J Cell Physiol ; 234(8): 12865-12875, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30537127

RESUMO

The balance between Ang II/AT1R and Ang-(1-7)/Mas plays a pivotal role in the development of lipopolysaccharides (LPS)-induced acute respiratory distress syndrome. However, the mechanisms underlying the balancing process still remain unclear. Here we investigated the roles of nuclear factor (NF)-κB and p53 in regulating AT1R and Mas expression. The results demonstrated that Ang II pretreatment resulted in downregulation of Mas and upregulation of AT1R, phosphorylated p65, and apoptosis in LPS-treated Human pulmonary microvascular endothelial cells (HPMVECs), but had no effect on p53 expression. Lentiviral vector-mediated P65 knockdown, but not a P53 knockdown, reversed all these effects of Ang II. On the other hand, Ang-(1-7) pretreatment lead to an increased in Mas expression and a decrease in AT1R, p53, and phosphorylated p65 expressions with suppressed apoptosis in LPS-treated cells. P65 knockdown promoted the protein expression of both AT1R and Mas while inhibiting p53 expression. P53 knockdown, but not a p65 knockdown, reversed all these effects of Ang-(1-7). Interestingly, p65 overexpression upregulated p53 and AT1R but downregulated Mas. P53 knockdown activated p65. These results suggest that there is a two-way feedback regulation between AT1R and Mas receptor via the NF-kB p65/P53 pathway, which may play a key role in LPS-induced HPMVECs apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Angiotensina II/farmacologia , Células Endoteliais/metabolismo , Humanos , Pulmão/metabolismo , NF-kappa B/metabolismo , Substâncias Protetoras/farmacologia , Regulação para Cima/efeitos dos fármacos
4.
Lab Invest ; 99(12): 1770-1783, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31278346

RESUMO

Early pulmonary fibrosis is the leading cause of poor prognosis in patients with acute respiratory distress syndrome (ARDS). However, whether the renin-angiotensin system (RAS) can serve as a therapeutic target is unknown. In this study, an animal model of early pulmonary fibrosis was established via the LPS three-hit regimen. Afterwards, the animals were treated with intraperitoneal injections of Ang-(1-7), AVE0991, or A779 once per day for 20 days. The plasma and BALF AngII levels of the animals were increased, while there were no significant changes in Ang-(1-7) levels in lung tissue after LPS treatment. Furthermore, the AT1R protein levels were significantly increased and the Mas levels were significantly decreased on days 14 and 21. Administration of Ang-(1-7) downregulated LPS-induced AT1R mRNA expression, which was upregulated by A779. The expression of Mas mRNA responded in the opposite direction relative to AT1R. Moreover, LPS caused decreased levels of Mas and E-cadherin and increased AT1R, Vimentin, and Src phosphorylation levels. Ang-(1-7) or AVE0991 blocked these effects but was counteracted by A779 treatment. Our findings suggested that AngII and AT1R levels exhibit opposite dynamic trends during LPS-induced early pulmonary fibrosis, as do Ang-(1-7) and Mas. Ang-(1-7) exerts protective effects against early pulmonary fibrosis, mainly by regulating the balance between AngII and AT1R and between Ang-(1-7) and Mas and by inhibiting Src kinase activation.


Assuntos
Angiotensina II/análogos & derivados , Angiotensina I/uso terapêutico , Imidazóis/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Fibrose Pulmonar/tratamento farmacológico , Vasodilatadores/uso terapêutico , Angiotensina I/sangue , Angiotensina II/sangue , Angiotensina II/farmacologia , Angiotensina II/uso terapêutico , Animais , Líquido da Lavagem Broncoalveolar/química , Caderinas/metabolismo , Avaliação Pré-Clínica de Medicamentos , Imidazóis/farmacologia , Lipopolissacarídeos , Pulmão/metabolismo , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/farmacologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/agonistas , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Fibrose Pulmonar/sangue , Distribuição Aleatória , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Fator de Crescimento Transformador beta/sangue , Vimentina/metabolismo
5.
Lab Invest ; 98(3): 339-359, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29200203

RESUMO

Pulmonary fibrosis triggered during the early stage of acute respiratory distress syndrome (ARDS) contributes to poor prognosis in patients. However, whether microRNAs (miRNAs) can serve as therapeutic targets for early pulmonary fibrosis during ARDS is still largely unknown. In this study, we evaluated the effects and mechanisms of miR-200s and its targets ZEB1/2 in lung tissue. An early pulmonary fibrosis mouse model caused by ARDS was established via a lipopolysaccharide (LPS) three-hit regimen. Lentiviral packaged miR-200b/c cDNA or ZEB1/2 shRNA was intratracheally administered into the lungs of C57BL/6 mice 1 day before an LPS injection was administered. In vitro, following a 30-min pretreatment with miR-200b/c or SB203580/SIS3, RLE-6TN cells were stimulated by LPS or LPS + transforming growth factor-ß (TGF-ß) for 24 h. miR-200b/c and E-cadherin protein expression declined, whereas ZEB1/2 mRNA and protein and vimentin and α-smooth muscle actin (α-SMA) protein levels gradually increased during the development of pulmonary fibrosis. Furthermore, both the overexpression of miR-200b/c and the silencing of ZEB1/2 significantly alleviated pulmonary inflammation and fibrosis, reduced vimentin and α-SMA expression, and increased E-cadherin protein levels. In RLE-6TN cells, LPS combined with TGF-ß exerts synergistic effects of increasing vimentin and α-SMA protein levels, increasing p38 and smad3 phosphorylation and reducing E-cadherin protein levels, which were reversed by pretreatment with miR-200b/c or SB203580/SIS3. Our findings demonstrate that miR-200b/c was downregulated, whereas ZEB1/2 was upregulated in the development of LPS-induced early pulmonary fibrosis. miR-200b/c exerts a protective effect by targeting ZEB1/2, which may be associated with the inhibition of p38 MAPK and TGF-ß /smad3 signaling pathways.


Assuntos
Sistema de Sinalização das MAP Quinases , MicroRNAs/metabolismo , Fibrose Pulmonar/metabolismo , Homeobox 2 de Ligação a E-box com Dedos de Zinco/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo , Actinas/metabolismo , Animais , Líquido da Lavagem Broncoalveolar/química , Caderinas/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal , Lipopolissacarídeos , Pulmão/patologia , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/etiologia , Fibrose Pulmonar/patologia , RNA Interferente Pequeno , Distribuição Aleatória , Ratos , Síndrome do Desconforto Respiratório/complicações , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Vimentina/metabolismo
6.
J Surg Res ; 219: 325-333, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29078900

RESUMO

BACKGROUND: Potential of liver regeneration after living-donor liver transplantation is closely associated with the recipient's prognosis, whereas exogenous gene might regulate the liver regeneration progress. NM23 is a multifunctional gene, which inhibits tumor metastasis and regulates cell proliferation, differentiation, development, and apoptosis; however, there is little research about NM23 in promoting liver cell proliferation. METHODS: To investigate the effect of NM23-E2 on the liver cell proliferation, the NM23-E2 overexpression vector or negative control vector was transfected into BRL-3A cells and donor liver, respectively. NM23-E2, Cyclin D1, and PCNA expression levels in BRL-3A cells and liver tissues were detected by quantitative real-time polymerase chain reaction and Western blot analysis. Cell Counting Kit-8 was used to detect cell proliferation and flow cytometry for investigating cell cycle. The liver regeneration rate was determined by calculating (regenerated-liver weight of recipient - liver weight of donor/liver weight of donor) × 100%. RESULTS: NM23-E2 overexpression increased the NM23-E2, Cyclin D1, and PCNA levels significantly in BRL-3A cells and liver tissues (P < 0.05). The number of S phase cells was more than that of negative control group, and cell proliferation rate was higher than that of the control group in BRL-3A cells markedly (P < 0.05). Moreover, the liver regeneration rate in the NM23-E2 overexpression group was also higher than that in negative control group on postoperative day 1, day 3, day 5, and day 7. CONCLUSIONS: Overexpression of NM23-E2 can increase Cyclin D1 and PCNA expression, shorten cell cycle, and thereby promoting the proliferation of liver cells and accelerating the regeneration of liver after 40% decreased-size rat liver transplantation.


Assuntos
Terapia Genética , Regeneração Hepática , Transplante de Fígado , Nucleosídeo NM23 Difosfato Quinases/genética , Animais , Ciclo Celular , Linhagem Celular , Proliferação de Células , Ciclina D1/metabolismo , Lentivirus , Fígado/metabolismo , Masculino , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ratos Sprague-Dawley
7.
Apoptosis ; 21(1): 69-84, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26456506

RESUMO

Pulmonary arterial hypertension (PAH) is a life-threatening disorder that ultimately causes heart failure. While the underlying causes of this condition are not well understood, previous studies suggest that the anti-apoptotic nature of pulmonary microvascular endothelial cells (PMVECs) in hypoxic environments contributes to PAH pathogenesis. In this study, we focus on the contribution of Bcl-2 and hypoxia response element (HRE) to apoptosis-resistant endothelial cells and investigate the mechanism. PMVECs obtained from either normal rats or apoptosis-resistant PMVECs obtained from PAH rats were transduced with recombinant lentiviral vectors carrying either Bcl-2-shRNA or HRE combined Bcl-2-shRNA, and then cultured these cells for 24 h under hypoxic (5% O2) or normoxic (21% O2) conditions. In normal PMVECs, Bcl-2-shRNA or HRE combined with Bcl-2-shRNA transduction successfully decreased Bcl-2 expression, while increasing apoptosis as well as caspase-3 and P53 expression in a normoxic environment. In a hypoxic environment, the effects of Bcl-2-shRNA treatment on cell apoptosis, and on Bcl-2, caspase-3, P53 expression were significantly suppressed. Conversely, HRE activation combined with Bcl-2-shRNA transduction markedly enhanced cell apoptosis and upregulated caspase-3 and P53 expression, while decreasing Bcl-2 expression. Furthermore, in apoptosis-resistant PMVECs, HRE-mediated Bcl-2 silencing effectively enhanced cell apoptosis and caspase-3 activity. The apoptosis rate was significantly depressed when Lv-HRE-Bcl-2-shRNA was combined with Lv-P53-shRNA or Lv-caspase3-shRNA transduction in a hypoxic environment. These results suggest that HRE-mediated Bcl-2 inhibition can effectively attenuate hypoxia-induced apoptosis resistance in PMVECs by downregulating Bcl-2 expression and upregulating caspase-3 and P53 expression. This study therefore reveals critical insight into potential therapeutic targets for treating PAH.


Assuntos
Apoptose/genética , Células Endoteliais/metabolismo , Hipertensão/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Elementos de Resposta , Animais , Caspase 3/genética , Caspase 3/metabolismo , Hipóxia Celular , Células Endoteliais/patologia , Regulação da Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Hipertensão/patologia , Lentivirus/genética , Lentivirus/metabolismo , Pulmão/irrigação sanguínea , Pulmão/patologia , Masculino , Monocrotalina , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Transdução Genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
8.
Int J Clin Pharmacol Ther ; 54(12): 987-991, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27668696

RESUMO

PURPOSE: To evaluate the corrective effect of norepinephrine on hypotension induced by dexmedetomidine through monitoring sedation status, hemodynamics as well as oxygen metabolism. METHODS: 100 patients administered standard-dose dexmedetomidine therapy with RASS between -2 and 0 in the intensive care unit (ICU) were included in the study. According to the application of norepinephrine to correct hypotension after dexmedetomidine therapy, the patients were divided into two groups: group A and group B. Group A (dexmedetomidine + norepinephrine group): those who had a systolic arterial pressure < 90 mmHg, a mean arterial pressure < 70 mmHg, or a decline in systolic arterial pressure > 40 mmHg or more than 30% of its base value after dexmedetomidine therapy and then received additional norepinephrine intravenously in order to maintain the arterial pressure at its base value. Group B (dexmedetomidine group): patients received the equivalent dose of dexmedetomidine to maintain the pressure at normal value without extra vasoconstrictor substance. Sedation (CPOT and RASS) and hemodynamic and oxygen metabolism indexes (heart rate, mean arterial pressure, respiratory rate, arterial oxygen saturation, central venous pressure, venous oxygen saturation, arteriovenous carbon dioxide difference, blood lactate level, blood lactate clearance rate, and average hourly urine output) were evaluated in the two groups at baseline, 6th hour, 12th hour, and 24th hour after the administration of intravenous dexmedetomidine. RESULTS: 39 cases were enrolled in group A and 61 cases in group B. Patients of both groups received adequate analgesia and sedation, and there was no significant statistical difference in analgesia and sedation at any point (both p > 0.05). Basal hemodynamic indexes and oxygen metabolism indexes also had no significant statistical difference (both p > 0.05). Central venous pressure (CVP) of group A was significantly higher than that of group B at the 6th hour and 12th hour after administration of intravenous dexmedetomidine (p = 0.005), and the heart rate (HR) of group A was markedly higher than that of group B at the 24th hour after dexmedetomidine therapy (p = 0.017), while the other indexes had no significant difference at any point (both p > 0.05). CONCLUSION: Dexmedetomidine plays an important role in ICU patients due to its pharmacological ability of sedation and analgesia. In our study, dexmedetomidine was successfully applied to ensure goal-directed sedation therapy (GDST). Norepinephrine can correct hypotension and bradycardia induced by intravenous dexmedetomidine. According to the hemodynamic indexes and oxygen metabolism indexes, the application of dexmedetomidine or the combination of dexmedetomidine with norepinephrine are both safe and appropriate to maintain the sedation status and hemodynamic situation in ICU patients.
.


Assuntos
Dexmedetomidina/efeitos adversos , Hipotensão/tratamento farmacológico , Norepinefrina/uso terapêutico , Idoso , Estado Terminal , Feminino , Hemodinâmica/efeitos dos fármacos , Humanos , Hipotensão/induzido quimicamente , Masculino , Pessoa de Meia-Idade
9.
J Nanosci Nanotechnol ; 15(2): 1821-30, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26353738

RESUMO

SWNHs induces accumulate with the levels of cytotoxic effects on different cell types and organs in animal models; it in a wide range may used for biomedical imaging. Conjunctival melanoma is a rare but potentially fatal ocular surface tumor. Recent studies of us have indicated the ability of SWNHs to inhibit proliferation of conjunctival melanoma cell line CRMM-1. But the role and molecular mechanisms of it was unclear. To address this question, the research about direct role of SWNHs on the growth, proliferation and apoptosis of CRMM-1 had been performed. Our results indicate that followed with the increasing concentrations of SWNHs, the number of cells decreased and apoptotic cells increased significantly. SWNHs delayed obviously mitotic entry of cells, and these effects followed the cultured time and the gradually increasing concentrations of SWNHs. SWNHs inhibited proliferation of cells at each time point in a time and dose-dependent manner, too. The TEM images showed that individual spherical SWNHs particles smaller than 100 nm in diameters were localized inside cytoplasma of cells. In summary, SWNHs inhibited mitotic entry, growth and proliferation of conjunctival melanoma cells and promoted its apoptosis, inhibited energy metabolism of cells in a dose-dependent manner. The roles of SWNHS on conjunctival melanoma cells were implicating energy metabolism. It may be the effective methods for treatment to conjunctival melanoma.


Assuntos
Carbono/administração & dosagem , Neoplasias da Túnica Conjuntiva/tratamento farmacológico , Neoplasias da Túnica Conjuntiva/metabolismo , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Nanopartículas/administração & dosagem , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Carbono/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Teste de Materiais , Nanopartículas/química , Nanopartículas/ultraestrutura , Consumo de Oxigênio/efeitos dos fármacos , Tamanho da Partícula , Resultado do Tratamento
10.
Mol Med Rep ; 29(1)2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38038164

RESUMO

Following the publication of this paper, it was drawn to the Editor's attention by a concerned reader that the ß­actin control western blotting data shown in Fig. 3D on p. 1893 were very similar to the contol data shown in Fig. 4A on p. 1894; furthermore, the data shown for the MMP­9 and the INOS protein bands in Fig. 4C were remarkably similar to the data shown for the IL­1ß and IL­6 proteins, respectively, albeit the backgrounds surrounding the bands were different. Moreover, various of the western blotting data shown in these figures were strikingly similar to data that had already been published in different form in other articles written by (largely) different authors at different research institutes. Owing to the fact that the contentious data in the above article had already been published prior to its submission to Molecular Medicine Reports, and due to the number of apparent duplications of strikingly similar data between Figs. 3 and 4, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a satisfactory reply. The Editor apologizes to the readership for any inconvenience caused. [Molecular Medicine Reports 7: 1889­1895, 2013; DOI: 10.3892/mmr.2013.1444].

11.
Int J Nanomedicine ; 18: 7745-7758, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38144514

RESUMO

Introduction: The incidence and mortality rates of sepsis-induced acute kidney injury (SAKI) remain high, posing a substantial healthcare burden. Studies have implicated a connection between the development of SAKI and inflammation response, apoptosis, and autophagy. Moreover, evidence suggests that manipulating autophagy could potentially influence the prognosis of this condition. Notably, exosomes derived from bone mesenchymal stem cells (BMSCs-Exo) have exhibited promise in mitigating cellular damage by modulating pathways associated with inflammation, apoptosis, and autophagy. Thus, this study aims to investigate the influence of BMSCs-Exo on SAKI and the potential mechanisms that drive this impact. Methods: The SAKI model was induced in HK-2 cells using lipopolysaccharide (LPS), while rats underwent cecal ligation and puncture (CLP) to simulate the condition. Cell viability was assessed using the CCK-8 kit, and kidney damage was evaluated through HE staining, blood urea nitrogen (BUN), and serum creatinine (SCr) measurements. Inflammatory-related RNAs and proteins were quantified via qPCR and ELISA, respectively. Apoptosis was determined through apoptosis-related protein levels, flow cytometry, and TUNEL staining. Western blot analysis was utilized to measure associated protein expressions. Results: In vivo, BMSCs-Exo ameliorated kidney injury in CLP-induced SAKI rats, reducing inflammatory cytokine production and apoptosis levels. Fluorescence microscope observed the absorption of BMSCs-Exo by renal cells following injection via tail vein. In the SAKI rat kidney tissue, there was an upregulation of LC3-II/LC3-I, p62, and phosphorylated AMP-activated protein kinase (p-AMPK) expressions, indicating blocked autophagic flux, while phosphorylated mammalian target of rapamycin (p-mTOR) expression was downregulated. However, BMSCs-Exo enhanced LC3-II/LC3-I and p-AMPK expression, concurrently reducing p62 and p-mTOR levels. In vitro, BMSCs-Exo enhanced cell viability in LPS-treated HK-2 cells, and exerted anti-inflammation and anti-apoptosis effects which were consistent with the results in vivo. Similarly, rapamycin (Rapa) exhibited a protective effect comparable to BMSCs-Exo, albeit partially abrogated by 3-methyladenine (3-MA). Conclusion: BMSCs-Exo mitigate inflammation and apoptosis through autophagy in SAKI, offering a promising avenue for SAKI treatment.


Assuntos
Injúria Renal Aguda , Exossomos , Células-Tronco Mesenquimais , Sepse , Ratos , Animais , Exossomos/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Quinases Ativadas por AMP/farmacologia , Lipopolissacarídeos/farmacologia , Células-Tronco Mesenquimais/metabolismo , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/terapia , Apoptose , Sepse/complicações , Sepse/terapia , Sepse/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Mamíferos
12.
BMC Mol Cell Biol ; 24(1): 2, 2023 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-36658496

RESUMO

Autophagy and pyroptosis of macrophages play important protective or detrimental roles in sepsis. However, the underlying mechanisms remain unclear. High mobility group box protein 1 (HMGB1) is associated with both pyroptosis and autophagy. lipopolysaccharide (LPS) is an important pathogenic factor involved in sepsis. Lentivirus-mediated HMGB1 shRNA was used to inhibit the expression of HMGB1. Macrophages were treated with acetylation inhibitor (AA) to suppress the translocation of HMGB1 from the nucleus to the cytosol. Autophagy and pyroptosis-related protein expressions were detected by Western blot. The levels of caspase-1 activity were detected and the rate of pyroptotic cells was detected by flow cytometry. LPS induced autophagy and pyroptosis of macrophages at different stages, and HMGB1 downregulation decreased LPS-induced autophagy and pyroptosis. Treatment with acetylation inhibitor (anacardic acid) significantly suppressed LPS-induced autophagy, an effect that was not reversed by exogenous HMGB1, suggesting that cytoplasmic HMGB1 mediates LPS-induced autophagy of macrophages. Anacardic acid or an anti-HMGB1 antibody inhibited LPS-induced pyroptosis of macrophages. HMGB1 alone induced pyroptosis of macrophages and this effect was inhibited by anti-HMGB1 antibody, suggesting that extracellular HMGB1 induces macrophage pyroptosis and mediates LPS-induced pyroptosis. In summary, HMGB1 plays different roles in mediating LPS-induced autophagy and triggering pyroptosis according to subcellular localization.


Assuntos
Proteína HMGB1 , Macrófagos , Sepse , Autofagia , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo , Macrófagos/metabolismo , Piroptose , Sepse/metabolismo , Animais
13.
Hum Cell ; 36(2): 568-582, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36478088

RESUMO

Patients with sepsis-induced acute respiratory distress syndrome (ARDS) have higher mortality and poor prognosis than pneumonia-induced ARDS. Pulmonary fibrosis is an irreversible accumulation of connective tissue in the interstitium of the lung and closely associated with the epithelial-mesenchymal transition (EMT) of type II alveolar epithelial cells (AECIIs). Therefore, it is undoubtedly worth studying whether the EMT of AECIIs in sepsis-induced ARDS patients is different from that in patients with pneumonia-induced ARDS in the regulatory mechanism. Here, we will report for the first time that an lncRNA-ASLNC12002 is highly expressed in AECIIs of patients with sepsis-induced pneumonia and promotes EMT in AECIIs. The research results showed that the expression of ASLNC12002 in AECIIs derived from patients with sepsis-induced ARDS is significantly higher than that in normal people and pneumonia-induced ARDS patients. Mechanism research showed that ASLNC12002 can cause the inactivation of the anti-EMT pathway NR2F2/miR128-3p/Snail1 by acting as the sponge of miR128-3p. Functional experiments showed that targeted silencing of ASLNC12002 could effectively inhibit EMT progression in AECIIs of patients with sepsis-induced pneumonia by restoring NR2F2/miR128-3p/Snail1 pathway. In a word, our study shows for the first time that the inactivation of NR2F2/miR128-3p/Snail1 pathway caused by the enhanced expression of ASLNC12002 is the direct reason why AECIIs in sepsis-induced ARDS patients are prone to get EMT progress. ASLNC12002 has the potential to become a biological target for the prevention and treatment of pulmonary fibrosis in patients with sepsis-induced ARDS. At the same time, the expectation that ASLNC12002 and its related products may be used as clinical markers for the evaluation of early pulmonary fibrosis in ARDS patients should not be ignored.


Assuntos
Pneumonia , Fibrose Pulmonar , RNA Longo não Codificante , Síndrome do Desconforto Respiratório , Sepse , Humanos , Células Epiteliais Alveolares/metabolismo , Fibrose Pulmonar/metabolismo , RNA Longo não Codificante/metabolismo , Transição Epitelial-Mesenquimal , Pneumonia/metabolismo
14.
Biomed Res Int ; 2021: 5513886, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34988226

RESUMO

BACKGROUND: Postoperative cognitive dysfunction (POCD) is a common complication after abdominal surgery. Several studies have reported that POCD is related to neuroinflammation caused by surgery. Omega-3 polyunsaturated fatty acids (PUFAs) can effectively inhibit the systematic inflammatory response. So, we use fish oil to study the effect of fish oil on inflammation, immunity, and cognitive behavior after splenectomy in rats. METHODS: 60 SD (Sprague-Dawley) rats were randomly divided into control group (group C, n = 20), surgery group (group S, n = 20), and omega-3 (fish oil) intervention group (group F, n = 20). Omega-3 PUFA was injected intraperitoneally from 3 days before operation to 7 days after operation in group F, and normal saline was injected simultaneously in group S. Rats in group S and group F received splenectomy under general anesthesia. Morris water maze behavioral evaluation was performed on the first, third, fifth, and seventh day after operation. The levels of IL-1ß (interleukin-1ß), IL-6 (interleukin-6), TNF-α (tumor necrosis factor-α), SOD (superoxide dismutase), and GSH-PX (glutathione peroxidase) were detected. RESULTS: Serums IL-1ß, IL-6, and TNF-α concentrations in group S and group F were higher than those in group C (P < 0.01), while those inflammatory cytokines in group F were significantly lower than those in group S (P < 0.01); serum GSH-PX levels in group F were higher than group S (P < 0.01). The Morris water maze behavior test performance of group F was better than that of group S (P < 0.05). CONCLUSION: Omega-3 PUFA can effectively improve postoperative inflammatory response, reduce the damage of antioxidant defense system, and improve postoperative cognitive function.


Assuntos
Cognição/efeitos dos fármacos , Ácidos Graxos Ômega-3/administração & dosagem , Animais , Antioxidantes/metabolismo , Óleos de Peixe/administração & dosagem , Glutationa Peroxidase/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Inflamação/metabolismo , Inflamação/fisiopatologia , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Aprendizagem em Labirinto/fisiologia , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Esplenectomia/métodos , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
15.
Cell Death Dis ; 12(7): 685, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34238924

RESUMO

Acute lung injury induced by ischemia-reperfusion (I/R)-associated pulmonary inflammation is associated with high rates of morbidity. Despite advances in the clinical management of lung disease, molecular therapeutic options for I/R-associated lung injury are limited. Zinc finger protein 36 (ZFP36) is an AU-rich element-binding protein that is known to suppress the inflammatory response. A ZFP36 binding site occurs in the 3' UTR of the cAMP-response element-binding protein (CREB) binding protein (CREBBP) gene, which is known to interact with apoptotic proteins to promote apoptosis. In this study, we investigate the involvement of ZFP36 and CREBBP on I/R-induced lung injury in vivo and in vitro. Intestinal ischemia/reperfusion (I/R) activates inflammatory responses, resulting in injury to different organs including the lung. Lung tissues from ZFP36-knockdown mice and mouse lung epithelial (MLE)-2 cells were subjected to either Intestinal I/R or hypoxia/reperfusion, respectively, and then analyzed by Western blotting, immunohistochemistry, and real-time PCR. Silico analyses, pull down and RIP assays were used to analyze the relationship between ZFP36 and CREBBP. ZFP36 deficiency upregulated CREBBP, enhanced I/R-induced lung injury, apoptosis, and inflammation, and increased I/R-induced lung fibrosis. In silico analyses indicated that ZFP36 was a strong negative regulator of CREBBP mRNA stability. Results of pull down and RIP assays confirmed that ZFP36 direct interacted with CREBBP mRNA. Our results indicated that ZFP36 can mediate the level of inflammation-associated lung damage following I/R via interactions with the CREBBP/p53/p21/Bax pathway. The downregulation of ZFP36 increased the level of fibrosis.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Proteína de Ligação a CREB/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Intestinos/irrigação sanguínea , Pulmão/metabolismo , Fibrose Pulmonar/prevenção & controle , Traumatismo por Reperfusão/complicações , Tristetraprolina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2/metabolismo , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Apoptose , Proteína de Ligação a CREB/genética , Linhagem Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal , Mediadores da Inflamação/metabolismo , Pulmão/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibrose Pulmonar/etiologia , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Transdução de Sinais , Tristetraprolina/genética
16.
Cell Death Dis ; 12(6): 614, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34131106

RESUMO

Among several leading cardiovascular disorders, ischemia-reperfusion (I/R) injury causes severe manifestations including acute heart failure and systemic dysfunction. Recently, there has been increasing evidence suggesting that alterations in mitochondrial morphology and dysfunction also play an important role in the prognosis of cardiac disorders. Long non-coding RNAs (lncRNAs) form major regulatory networks altering gene transcription and translation. While the role of lncRNAs has been extensively studied in cancer and tumor biology, their implications on mitochondrial morphology and functions remain to be elucidated. In this study, the functional roles of Zinc finger protein 36-like 2 (ZFP36L2) and lncRNA PVT1 were determined in cardiomyocytes under hypoxia/reoxygenation (H/R) injury in vitro and myocardial I/R injury in vivo. Western blot and qRT-PCR analysis were used to assess the levels of ZFP36L2, mitochondrial fission and fusion markers in the myocardial tissues and cardiomyocytes. Cardiac function was determined by immunohistochemistry, H&E staining, and echocardiogram. Ultrastructural analysis of mitochondrial fission was performed using transmission electron microscopy. The mechanistic model consisting of PVT1 with ZFP36L2 and microRNA miR-21-5p with E3 ubiquitin ligase MARCH5 was assessed by subcellular fraction, RNA pull down, FISH, and luciferase reporter assays. These results identified a novel regulatory axis involving PVT1, miR-21-5p, and MARCH5 that alters mitochondrial morphology and function during myocardial I/R injury. Using an in vivo I/R injury mouse model and in vitro cardiomyocytes H/R model, we demonstrated that ZFP36L2 directly associates with PVT1 and alters mitochondrial fission and fusion. PVT1 also interactes with miR-21-5p and suppresses its expression and activity. Furthermore, we identified MARCH5 as a modifier of miR-21-5p, and its effect on mitochondrial fission and fusion are directly proportional to PVT1 expression during H/R injury. Our findings show that manipulation of PVT1-miR-21-5p-MARCH5-mediated mitochondrial fission and fusion via ZFP36L2 may be a novel therapeutic approach to regulate myocardial I/R injury.


Assuntos
Dinâmica Mitocondrial/genética , Traumatismo por Reperfusão Miocárdica/genética , RNA Longo não Codificante/fisiologia , Tristetraprolina/fisiologia , Animais , Células Cultivadas , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia
17.
Oxid Med Cell Longev ; 2021: 6628847, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33981387

RESUMO

Sepsis-induced acute kidney injury (SI-AKI) is a serious condition in critically ill patients. Currently, the diagnosis is based on either elevated serum creatinine levels or oliguria, which partially contribute to delayed recognition of AKI. Metabolomics is a potential approach for identifying small molecule biomarkers of kidney diseases. Here, we studied serum metabolomics alterations in rats with sepsis to identify early biomarkers of sepsis and SI-AKI. A rat model of SI-AKI was established by intraperitoneal injection of lipopolysaccharide (LPS). Thirty Sprague-Dawley (SD) rats were randomly divided into the control (CT) group and groups treated for 2 hours (LPS2) and 6 hours (LPS6) with LPS (10 rats per group). Nontargeted metabolomics screening was performed on the serum samples from the control and SI-AKI groups. Combined multivariate and univariate analysis was used for pairwise comparison of all groups to identify significantly altered serum metabolite levels in early-stage AKI in rats with sepsis. Orthogonal partial least squares discriminant analysis (OPLS-DA) showed obvious separation between the CT and LPS2 groups, CT and LPS6 groups, and LPS2 and LPS6 groups. All comparisons of the groups identified a series of differential metabolites according to the threshold defined for potential biomarkers. Intersections and summaries of these differential metabolites were used for pathway enrichment analysis. The results suggested that sepsis can cause an increase in systemic aerobic and anaerobic metabolism, an impairment of the oxygen supply, and uptake and abnormal fatty acid metabolism. Changes in the levels of malic acid, methionine sulfoxide, and petroselinic acid were consistently measured during the progression of sepsis. The development of sepsis was accompanied by the development of AKI, and these metabolic disorders are directly or indirectly related to the development of SI-AKI.


Assuntos
Injúria Renal Aguda/etiologia , Biomarcadores/química , Metabolômica/métodos , Sepse/complicações , Humanos
18.
Cell Death Differ ; 27(9): 2635-2650, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32203170

RESUMO

Acute lung injury (ALI) is a life-threatening disorder with high rates of morbidity and mortality. Reactive oxygen species and epithelial apoptosis are involved in the pathogenesis of acute lung injury. Ferroptosis, an iron-dependent non-apoptotic form of cell death, mediates its effects in part by promoting the accumulation of reactive oxygen species. The inhibition of ferroptosis decreases clinical symptoms in experimental models of ischemia/reperfusion-induced renal failure and heart injury. This study investigated the roles of inhibitor of apoptosis-stimulating protein of p53 (iASPP) and Nrf2 in ferroptosis and their potential therapeutic effects in intestinal ischemia/reperfusion-induced acute lung injury. Intestinal ischemia/reperfusion-induced ALI was induced in wild-type and Nrf2-/- mice. The mice were treated with erastin followed by liproxstatin-1. Ferroptosis-related factors in mice with ischemia/reperfusion-induced acute lung injury or in mouse lung epithelial-2 cells with hypoxia/regeneration (HR)-induced ALI were measured by western blotting, real-time PCR, and immunofluorescence. Ferroptosis contributed to intestinal ischemia/reperfusion-induced ALI in vivo. iASPP inhibited ferroptosis and alleviated intestinal ischemia/reperfusion-induced acute lung injury, and iASPP-mediated protection against ischemia/reperfusion-induced ALI was dependent on Nrf2 signaling. HR-induced acute lung injury enhanced ferroptosis in vitro in mouse lung epithelial-2 cells, and ferroptosis was modulated after the enhancement of intestinal ischemia/reperfusion in Nrf2-/- mice. iASPP mediated its protective effects against acute lung injury through the Nrf2/HIF-1/TF signaling pathway. Ferroptosis contributes to intestinal ischemia/reperfusion-induced ALI, and iASPP treatment inhibits ferroptosis in part via Nrf2. These findings indicate the therapeutic potential of iASPP for treating ischemia/reperfusion-induced ALI.


Assuntos
Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/metabolismo , Ferroptose , Intestinos/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Traumatismo por Reperfusão/complicações , Proteínas Repressoras/metabolismo , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/prevenção & controle , Animais , Ferroptose/efeitos dos fármacos , Hipóxia/complicações , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 2 Relacionado a NF-E2/deficiência , Fator 2 Relacionado a NF-E2/metabolismo , Piperazinas/farmacologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos
19.
Front Mol Biosci ; 6: 152, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31921892

RESUMO

Sepsis-induced acute kidney injury (AKI) can increase the mortality of critically ill patients and the incidence of chronic kidney disease in critically ill survivors. The main goal was to investigate the possible link between metabolic changes and sepsis-induced AKI development. The experimental animal model of sepsis-induced AKI was established by intraperitoneal injection of lipopolysaccharide in rats. Non-targeted metabolic screening of the renal cortex in the control and sepsis-induced AKI groups was carried out based on gas chromatography coupled with quadrupole time-of-flight mass spectrometry (GC-TOFMS) technology. The data between the two groups were analyzed by combining univariate and multivariate statistical methods, and the metabolites associated with AKI in rats with sepsis were screened. By examining the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, altered metabolic pathways associated with acute renal injury in sepsis were identified. The cross validated scores plot of orthogonal partial least squares discriminant analysis (OPLS-DA) showed a distinct separation trend between the model and control groups in the profile of renal cortex metabolites, indicating a significant change in endogenous metabolites in the rat renal cortex. Further analysis and screening showed that 26 different metabolites were identified in the renal cortex between the two groups, mainly involving taurine and hypotaurine metabolism, pantothenic acid and CoA biosynthesis, phenylalanine metabolism, and other metabolic pathways. The metabolic disorders of taurine, pantothenic acid, and phenylalanine in the renal cortex are related to the development of acute renal injury in sepsis. Correcting these metabolic disorders is expected to prevent and treat sepsis-induced AKI.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA