Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Transl Res ; 270: 66-80, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38604333

RESUMEN

Sepsis-induced acute lung injury (ALI) is a serious complication of sepsis and the predominant cause of death. Exosomes released by lung tissue cells critically influence the progression of ALI during sepsis by modulating the inflammatory microenvironment. However, the molecular mechanisms by which exosome-mediated intercellular signaling exacerbates ALI in septic infection remain undefined. Our study found increased levels of exosomal Tenascin-C (TNC) in the plasma of both patients and mice with ALI, showing a strong association with disease progression. By integrating exosomal proteomics with transcriptome sequencing and experimental validation, we elucidated that LPS induce unresolved endoplasmic reticulum stress (ERs) in alveolar epithelial cells (AECs), ultimately leading to the release of exosomal TNC through the activation of PERK-eIF2α and the transcription factor CHOP. In the sepsis mouse model with TNC knockout, we noted a marked reduction in macrophage pyroptosis. Our detailed investigations found that exosomal TNC binds to TLR4 on macrophages, resulting in an augmented production of ROS, subsequent mitochondrial damage, activation of the NF-κB signaling pathway, and induction of DNA damage response. These interconnected events culminate in macrophage pyroptosis, thereby amplifying the release of inflammatory cytokines. Our findings demonstrate that exosomal Tenascin-C, released from AECs under unresolved ER stress, exacerbates acute lung injury by intensifying sepsis-associated inflammatory responses. This research provides new insights into the complex cellular interactions underlying sepsis-induced ALI.

2.
Front Pharmacol ; 15: 1261772, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38584603

RESUMEN

Introduction: Patients with sepsis are at an incremental risk of acute lung injury (ALI). Baiqian, also known as Cynanchi stauntonii rhizoma et radix (Csrer), has anti-inflammatory properties and is traditionally used to treat cough and phlegm. This study aimed to demonstrate the multicomponent, multitarget, and multi-pathway regulatory molecular mechanisms of Csrer in treating lipopolysaccharide (LPS)-induced ALI. Methods: The bioactive components of Csrer were identified by ultrahigh-performance liquid chromatography Q-Orbitrap mass spectrometry (UPLC-Q-Orbitrap MS). Active targets predicted from PharmMapper. DrugBank, OMIM, TTD, and GeneCards were used to identify potential targets related to ALI. Intersection genes were identified for Csrer against ALI. The PPI network was analysed to identify prime targets. GO and KEGG analyses were performed. A drug-compound-target-pathway-disease network was constructed. Molecular docking and simulations evaluated the binding free energy between key proteins and active compounds. The protective effect and mechanism of Csrer in ALI were verified using an ALI model in mice. Western blot, Immunohistochemistry and TUNEL staining evaluated the mechanisms of the pulmonary protective effects of Csrer. Results: Forty-six bioactive components, one hundred and ninety-two potential cross-targets against ALI and ten core genes were identified. According to GO and KEGG analyses, the PI3K-Akt, apoptosis and p53 pathways are predominantly involved in the "Csrer-ALI" network. According to molecular docking and dynamics simulations, ten key genes were firmly bound by the principal active components of Csrer. The "Csrer-ALI" network was revealed to be mediated by the p53-mediated apoptosis and inflammatory pathways in animal experiments. Conclusion: Csrer is a reliable source for ALI treatment based on its practical components, potential targets and pathways.

3.
Mil Med Res ; 11(1): 24, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38644472

RESUMEN

Sepsis, a severe systemic inflammatory response to infection, remains a leading cause of morbidity and mortality worldwide. Exosomes, as mediators of intercellular communication, play a pivotal role in the pathogenesis of sepsis through modulating immune responses, metabolic reprogramming, coagulopathy, and organ dysfunction. This review highlights the emerging significance of exosomes in these processes. Initially, it provides an in-depth insight into exosome biogenesis and characterization, laying the groundwork for understanding their diverse and intricate functions. Subsequently, it explores the regulatory roles of exosomes in various immune cells such as neutrophils, macrophages, dendritic cells, T cells, and B cells. This analysis elucidates how exosomes are pivotal in modulating immune responses, thus contributing to the complexity of sepsis pathophysiology. Additionally, this review delves into the role of exosomes in the regulation of metabolism and subsequent organ dysfunction in sepsis. It also establishes a connection between exosomes and the coagulation cascade, which affects endothelial integrity and promotes thrombogenesis in sepsis. Moreover, the review discusses the dual role of exosomes in the progression and resolution of sepsis, exploring their complex involvement in inflammation and healing processes. Furthermore, it underscores their potential as biomarkers and therapeutic targets. Understanding these mechanisms presents new opportunities for novel interventions to mitigate the severe outcomes of sepsis, emphasizing the therapeutic promise of exosome research in critical care settings.


Asunto(s)
Exosomas , Insuficiencia Multiorgánica , Sepsis , Exosomas/metabolismo , Humanos , Sepsis/fisiopatología , Sepsis/complicaciones , Sepsis/metabolismo , Insuficiencia Multiorgánica/fisiopatología , Insuficiencia Multiorgánica/etiología , Comunicación Celular/fisiología , Inflamación/fisiopatología , Animales
4.
Cell Rep ; 43(3): 113905, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38446660

RESUMEN

Motivation-driven mating is a basic affair for the maintenance of species. However, the underlying molecular mechanisms that control mating motivation are not fully understood. Here, we report that NRG1-ErbB4 signaling in the medial amygdala (MeA) is pivotal in regulating mating motivation. NRG1 expression in the MeA negatively correlates with the mating motivation levels in adult male mice. Local injection and knockdown of MeA NRG1 reduce and promote mating motivation, respectively. Consistently, knockdown of MeA ErbB4, a major receptor for NRG1, and genetic inactivation of its kinase both promote mating motivation. ErbB4 deletion decreases neuronal excitability, whereas chemogenetic manipulations of ErbB4-positive neuronal activities bidirectionally modulate mating motivation. We also identify that the effects of NRG1-ErbB4 signaling on neuronal excitability and mating motivation rely on hyperpolarization-activated cyclic nucleotide-gated channel 3. This study reveals a critical molecular mechanism for regulating mating motivation in adult male mice.


Asunto(s)
Motivación , Transducción de Señal , Ratones , Masculino , Animales , Neuronas/metabolismo , Receptor ErbB-4/metabolismo , Amígdala del Cerebelo/metabolismo , Neurregulina-1/metabolismo
5.
J Adv Res ; 44: 39-51, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35618635

RESUMEN

INTRODUCTION: Sepsis-induced apoptosis leads to lymphopenia including the decrease of CD4+ T cells thus favoring immunosuppression. OBJECTIVES: Although epidermal growth factor receptor (EGFR) inhibitors significantly improve the survival rate of septic mice, the effect of EGFR on the function and metabolism of CD4+ T cells in sepsis remained unknown. METHODS: CD4+ T cells from septic mice and patients were assessed for apoptosis, activation, Warburg metabolism and glucose transporter 1 (Glut1) expression with or without the interference of EGFR activation. RESULTS: EGFR facilitates CD4+ T cell activation and apoptosis through Glut1, which is a key enzyme that controls glycolysis in T cells. EGFR, TANK binding kinase 1 (TBK1) and Glut1 form a complex to facilitate Glut1 transportation from cytoplasm to cell surface. Both the levels of membrane expression of EGFR and Glut1 and the activation levels of CD4+ T cells were significantly higher in patients with sepsis as compared with healthy subjects. CONCLUSION: Our data demonstrated that through its downstream TBK1/Exo84/RalA protein system, EGFR regulates Glut1 transporting to the cell surface, which is a key step for inducing the Warburg effect and the subsequent cellular activation and apoptosis of CD4+ T lymphocytes and may eventually affect the immune functional status, causing immune cell exhaustion in sepsis.


Asunto(s)
Linfocitos T CD4-Positivos , Sepsis , Animales , Ratones , Linfocitos T CD4-Positivos/metabolismo , Receptores ErbB/metabolismo , Receptores ErbB/farmacología , Apoptosis , Sepsis/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo
6.
Cell Death Dis ; 13(11): 934, 2022 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-36344490

RESUMEN

EGFR phosphorylation is required for TLR4-mediated macrophage activation during sepsis. However, whether and how intracellular EGFR is transported during endotoxemia have largely been unknown. Here, we show that LPS promotes high levels cell surface expression of EGFR in macrophages through two different transport mechanisms. On one hand, Rab10 is required for EEA1-mediated the membrane translocation of EGFR from the Golgi. On the other hand, EGFR phosphorylation prevents its endocytosis in a kinase activity-dependent manner. Erlotinib, an EGFR tyrosine kinase inhibitor, significantly reduced membrane EGFR expression in LPS-activated macrophage. Mechanistically, upon LPS induced TLR4/EGFR phosphorylation, MAPK14 phosphorylated Rab7a at S72 impaired membrane receptor late endocytosis, which maintains EGFR membrane localization though blocking its lysosomal degradation. Meanwhile, Rab5a is also involved in the early endocytosis of EGFR. Subsequently, inhibition of EGFR phosphorylation switches M1 phenotype to M2 phenotype and alleviates sepsis-induced acute lung injury. Mechanistic study demonstrated that Erlotinib suppressed glycolysis-dependent M1 polarization via PKM2/HIF-1ɑ pathway and promoted M2 polarization through up-regulating PPARγ induced glutamine metabolism. Collectively, our data elucidated a more in-depth mechanism of macrophages activation, and provided stronger evidence supporting EGFR as a potential therapeutic target for the treatment of sepsis.


Asunto(s)
Endotoxemia , Sepsis , Humanos , Fosforilación , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo , Clorhidrato de Erlotinib , Activación de Macrófagos , Receptor Toll-Like 4/metabolismo , Receptores ErbB/metabolismo , Proteínas Tirosina Quinasas/metabolismo
7.
Front Immunol ; 13: 995974, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36203606

RESUMEN

Background: Sepsis-induced apoptosis of immune cells leads to widespread depletion of key immune effector cells. Endoplasmic reticulum (ER) stress has been implicated in the apoptotic pathway, although little is known regarding its role in sepsis-related immune cell apoptosis. The aim of this study was to develop an ER stress-related prognostic and diagnostic signature for sepsis through bioinformatics and machine learning algorithms on the basis of the differentially expressed genes (DEGs) between healthy controls and sepsis patients. Methods: The transcriptomic datasets that include gene expression profiles of sepsis patients and healthy controls were downloaded from the GEO database. The immune-related endoplasmic reticulum stress hub genes associated with sepsis patients were identified using the new comprehensive machine learning algorithm and bioinformatics analysis which includes functional enrichment analyses, consensus clustering, weighted gene coexpression network analysis (WGCNA), and protein-protein interaction (PPI) network construction. Next, the diagnostic model was established by logistic regression and the molecular subtypes of sepsis were obtained based on the significant DEGs. Finally, the potential diagnostic markers of sepsis were screened among the significant DEGs, and validated in multiple datasets. Results: Significant differences in the type and abundance of infiltrating immune cell populations were observed between the healthy control and sepsis patients. The immune-related ER stress genes achieved strong stability and high accuracy in predicting sepsis patients. 10 genes were screened as potential diagnostic markers for sepsis among the significant DEGs, and were further validated in multiple datasets. In addition, higher expression levels of SCAMP5 mRNA and protein were observed in PBMCs isolated from sepsis patients than healthy donors (n = 5). Conclusions: We established a stable and accurate signature to evaluate the diagnosis of sepsis based on the machine learning algorithms and bioinformatics. SCAMP5 was preliminarily identified as a diagnostic marker of sepsis that may affect its progression by regulating ER stress.


Asunto(s)
Biología Computacional , Sepsis , Estrés del Retículo Endoplásmico/genética , Perfilación de la Expresión Génica , Humanos , Aprendizaje Automático , Proteínas de la Membrana/genética , ARN Mensajero , Sepsis/diagnóstico , Sepsis/genética
8.
J Pain Res ; 15: 1837-1844, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35789818

RESUMEN

Purpose: In recent years, patient-centered postoperative quality of recovery has gained attention. This study aimed to assess the influence of ultrasound-guided continuous fascia iliaca compartment block (CFICB) on early quality of recovery in elderly patients after total hip arthroplasty (THA) using the QoR-15 score. Patients and Methods: In this single-center, randomized, prospective study, 60 patients scheduled for unilateral THA were randomized to the CFICB or patient-controlled intravenous analgesia (PCIA) group. The primary outcome was the QoR-15 score. The secondary outcomes were pain score, number of patients requiring rescue analgesics, time of first postoperative ambulation, incidence of postoperative complications, Bromage score, and length of hospital stay. Results: The QoR-15 score was significantly higher in the CFICB group than in the PCIA group at 24 h (P < 0.001) after surgery. However, the QoR-15 score was not significantly different at 48 h (P = 0.074) between the two groups. Pain scores at rest and during movement were lower in the CFICB group than in the PCIA group at 12, 24, and 48 h postoperatively (P < 0.05). There was no difference in the number of patients requiring rescue analgesics, time of first postoperative ambulation, incidence of postoperative complications apart from dizziness, or length of hospital stay between the two groups. In addition, Bromage score of 1 point was reported by four patients in the CFICB group at 24 h (P = 0.048) after THA. Conclusion: In elderly patients following THA, CFICB improved the quality of recovery at 24 h and reduced pain scores compared with PCIA. The time of first postoperative ambulation and length of hospital stay were not significantly affected.

9.
Commun Biol ; 5(1): 543, 2022 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-35668098

RESUMEN

Sepsis-induced acute lung injury (ALI) is a serious sepsis complication and the prevailing cause of death. Circulating plasma exosomes might exert a key role in regulating intercellular communication between immunological and structural cells, as well as contributing to sepsis-related organ damage. However, the molecular mechanisms by which exosome-mediated intercellular signaling exacerbate ALI in septic infection remains undefined. Therefore, we investigated the effect of macrophage-derived exosomal APN/CD13 on the induction of epithelial cell necrosis. Exosomal APN/CD13 levels in the plasma of septic mice and patients with septic ALI were found to be higher. Furthermore, increased plasma exosomal APN/CD13 levels were associated with the severity of ALI and fatality in sepsis patients. We found remarkably high expression of APN/CD13 in exosomes secreted by LPS-stimulated macrophages. Moreover, c-Myc directly induced APN/CD13 expression and was packed into exosomes. Finally, exosomal APN/CD13 from macrophages regulated necroptosis of lung epithelial cells by binding to the cell surface receptor TLR4 to induce ROS generation, mitochondrial dysfunction and NF-κB activation. These results demonstrate that macrophage-secreted exosomal APN/CD13 can trigger epithelial cell necroptosis in an APN/CD13-dependent manner, which provides insight into the mechanism of epithelial cell functional disorder in sepsis-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda , Sepsis , Lesión Pulmonar Aguda/complicaciones , Animales , Antígenos CD13/farmacología , Células Epiteliales , Humanos , Pulmón , Macrófagos , Ratones , Necroptosis , Sepsis/complicaciones
10.
Front Immunol ; 13: 859231, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35634298

RESUMEN

Heat stroke (HS) is a severe condition characterized by increased morbidity and high mortality. Acute liver injury (ALI) is a well-documented complication of HS. The tumor suppressor p53 plays an important role in regulation of mitochondrial integrity and mitophagy in several forms of ALI. However, the role of p53-regulated mitophagy in HS-ALI remains unclear. In our study, we discovered the dynamic changes of mitophagy in hepatocytes and demonstrated the protective effects of mitophagy activation on HS-ALI. Pretreatment with 3-MA or Mdivi-1 significantly exacerbated ALI by inhibiting mitophagy in HS-ALI mice. Consistent with the animal HS-ALI model results, silencing Parkin aggravated mitochondrial damage and apoptosis by inhibiting mitophagy in HS-treated normal human liver cell line (LO2 cells). Moreover, we described an increase in the translocation of p53 from the nucleus to the cytoplasm, and cytosolic p53 binds to Parkin in LO2 cells following HS. p53 overexpression using a specific adenovirus or Tenovin-6 exacerbated HS-ALI through Parkin-dependent mitophagy both in vivo and in vitro, whereas inhibition of p53 using siRNA or PFT-α effectively reversed this process. Our results demonstrate that cytosolic p53 binds to Parkin and inhibits mitophagy by preventing Parkin's translocation from the cytosol to the mitochondria, which decreases mitophagy activation and leads to hepatocyte apoptosis in HS-ALI. Overall, pharmacologic induction of mitophagy by inhibiting p53 may be a promising therapeutic approach for HS-ALI treatment.


Asunto(s)
Golpe de Calor , Mitofagia , Animales , Citosol/metabolismo , Hígado/metabolismo , Ratones , Mitofagia/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
11.
Acta Biochim Biophys Sin (Shanghai) ; 53(3): 342-353, 2021 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-33521809

RESUMEN

Acute lung injury (ALI) is an inflammatory pulmonary disease that can easily develop into serious acute respiratory distress syndrome, which has high morbidity and mortality. However, the molecular mechanism of ALI remains unclear, and few molecular biomarkers for diagnosis and treatment have been identified. In this study, we aimed to identify novel molecular biomarkers using a bioinformatics approach. Gene expression data were obtained from the Gene Expression Omnibus database, co-expressed differentially expressed genes (CoDEGs) were identified using R software, and further functional enrichment analyses were conducted using the online tool Database for Annotation, Visualization, and Integrated Discovery. A protein-protein interaction network was established using the STRING database and Cytoscape software. Lipopolysaccharide (LPS)-induced ALI mouse model was constructed and verified. The hub genes were screened and validated in vivo. The transcription factors (TFs) and miRNAs associated with the hub genes were predicted using the NetworkAnalyst database. In total, 71 CoDEGs were screened and found to be mainly involved in the cytokine-cytokine receptor interactions, and the tumor necrosis factor and malaria signaling pathways. Animal experiments showed that the lung injury score, bronchoalveolar lavage fluid protein concentration, and wet-to-dry weight ratio were higher in the LPS group than those in the control group. Real-time polymerase chain reaction analysis indicated that most of the hub genes such as colony-stimulating factor 2 (Csf2) were overexpressed in the LPS group. A total of 20 TFs including nuclear respiratory factor 1 (NRF1) and two miRNAs were predicted to be regulators of the hub genes. In summary, Csf2 may serve as a novel diagnostic and therapeutic target for ALI. NRF1 and mmu-mir-122-5p may be key regulators in the development of ALI.


Asunto(s)
Lesión Pulmonar Aguda , Biología Computacional , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos , Mapas de Interacción de Proteínas , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/metabolismo , Animales , Modelos Animales de Enfermedad , Lipopolisacáridos/toxicidad , Masculino , Ratones
12.
Am J Transl Res ; 12(11): 7528-7541, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33312387

RESUMEN

MicroRNA-219-5p (miR-219-5p) is a key post-transcriptional regulator of gene expression that is known to regulate cancer progression, but its role in the context of hepatocellular carcinoma (HCC) remains to be fully elucidated. Herein, it was found that this miRNA functions as a tumor suppressor. Specifically, significant decreases in miR-219-5p expression were detected in HCC cells and patient serum samples relative to that found in the serum of 15 healthy people, and it was concluded that miR-219-5p overexpression was sufficient to impair HCC cell proliferation in vitro and vivo and migration in vitro. At the mechanistic level, it was found that miR-219-5p was able to suppress the expression of NEK6 (never in mitosis gene a-related kinase 6), thereby resulting in dysregulated ß-catenin/c-Myc-regulated gene expression. When NEK6 was overexpressed in HCC cells, this was sufficient to reverse the inhibitory impact of miR-219-5p on HCC cell proliferation both in vitro and vivo and metastasis in vitro. Bioinformatics analyses were also conducted, and both miR-219-5p and Nek6 were linked to disease progression in HCC patients with advanced disease. More importantly, the serum specimen data showed that reduced perioperative plasma miR-219-5p correlated significantly with increased risk of early recurrence after curative hepatectomy, whereas it was opposed to NEK6. Together, these findings highlight miR-219-5p as a potentially valuable diagnostic biomarker that can potentially be leveraged to improve clinical outcomes in HCC patients.

13.
J Immunol ; 205(3): 767-775, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32580932

RESUMEN

Receptor for advanced glycation end-products (RAGE) and TLR4 play an important role in the inflammatory response against High-mobility group box 1 protein (HMGB1), a late proinflammatory cytokine and a damage-associated molecular pattern. As cell surface receptors, both RAGE and TLR4 are constantly trafficking between the cytoplasm and plasma membrane. However, whether TLR4 is related to the intracellular transport of RAGE in HMGB1-induced inflammation remains unknown. In this study, we demonstrated that HMGB1 not only increased RAGE expression in both the cytoplasm and plasma membrane but also upregulated the expression of TLR4 in the plasma membrane. Knocking out of RAGE led to decreased MAPK activation, TLR4 cellular membrane expression, and corresponding inflammatory cytokine generation. Meanwhile, inhibiting MAPK activation also decreased TLR4 surface expression. These results indicated that HMGB1 may bind to cell surface RAGE receptors on the cell surface, leading to MAPK activation, thus promoting TLR4 translocation on the cell surface, but does not regulate its transcription and translation. In contrast, TLR4 can increase the transcription and translation of RAGE, which translocates to the cell surface and is able to bind to more HMGB1. The cell surface receptors TLR4 and RAGE bind to HMGB1, leading to the transcription and secretion of inflammatory cytokines. Finally, we also observed these results in the mice pseudofracture model, which is closely related to HMGB1-induced inflammatory response. All these results demonstrated that the interplay between RAGE and TLR4 are critical for HMGB1-induced inflammatory response.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Proteína HMGB1/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Receptor para Productos Finales de Glicación Avanzada/inmunología , Receptor Toll-Like 4/inmunología , Animales , Membrana Celular/genética , Membrana Celular/inmunología , Citoplasma/genética , Citoplasma/inmunología , Proteína HMGB1/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Ratones Noqueados , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor Toll-Like 4/genética , Transcripción Genética/inmunología
14.
Oxid Med Cell Longev ; 2020: 6579696, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32148654

RESUMEN

Mitophagy is involved in sepsis-induced acute lung injury (ALI). Bcl-2 family proteins play an important role in mitochondrial homeostasis. However, whether targeting Bcl-2 proteins (Bcl-2 and Bad) could influence mitophagy in ALI remains unclear. In this study, lipopolysaccharide (LPS) was used to induce injury in A549 cells and ALI in mice. LPS treatment resulted in elevated cell apoptosis, enhanced mitophagy, decreased Bcl-2 expression, increased Bad expression, and activation of PINK1/Parkin signaling in cells and lung tissues. Both Bcl-2 overexpression and Bad knockdown attenuated LPS-induced injury, inhibited cell apoptosis and mitophagy, and improved survival. Atg5 knockout (KO) inhibited LPS-induced cell apoptosis. Furthermore, Bcl-2 proteins regulated mitophagy by modulating the recruitment of Parkin from the cytoplasm to mitochondria via direct protein-protein interactions. These results were further confirmed in Park2 KO cells and Park2-/- mice. This is the first study to demonstrate that Bcl-2 proteins regulated mitophagy in LPS-induced ALI via modulating the PINK1/Parkin signaling pathway, promoting new insights into the mechanisms and investigation of therapeutic strategies for a septic patient with ALI.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Lipopolisacáridos/efectos adversos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ubiquitina-Proteína Ligasas/genética , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Mitofagia , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo
15.
Clin Exp Pharmacol Physiol ; 47(1): 127-134, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31429973

RESUMEN

The biological roles of intravenous anaesthetic propofol in cancer have been shown by various studies using cancer cell lines that represent differentiated cancer cells. However, the activities of propofol in cancer stem cells have not been elucidated. In this work, we examined the effects and mechanisms of propofol on acute myeloid leukaemia (AML) differentiated and CD34+ CD38- stem cells. We found that propofol inhibited growth, differentiation and self-renewal capabilities of AML stem cells regardless of cellular origin and genetic profiling. In addition, propofol inhibited the growth of AML differentiated cells. Propofol significantly induced apoptosis of AML differentiated but not CD34+ CD38- stem cells. We further found that propofol significantly augmented the efficacy of AML standard therapeutic drugs. Consistent with the previous findings, we showed that propofol suppressed the Akt/mTOR pathway in AML cells. We also found that propofol inhibited pathways important for stem cell maintenance and self-renewal, such as Wnt/ß-catenin. Overexpression of constitutively active Akt partially reversed the inhibitory effects of propofol in AML differentiated cells. Stabilization of ß-catenin using genetic and pharmacological approaches also partially rescued the inhibitory effects of propofol in AML differentiated and stem cells. Our work shows that propofol targets leukaemia cells at all stages of development, in a cell type-specific manner. Inhibition of both Akt/mTOR and Wnt/ß-catenin is required for the action of propofol in AML. Our findings also highlight the activities of propofol on cancer stem cells.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Propofol/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo , ADP-Ribosil Ciclasa 1/metabolismo , Antígenos CD34/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
16.
Clin Ther ; 41(9): 1864-1877.e0, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31345559

RESUMEN

PURPOSE: To investigate the optimal agent combined with propofol for sedation in elderly patients undergoing gastrointestinal endoscopy. METHODS: A total of 120 elderly patients scheduled for gastrointestinal endoscopy under propofol-based sedation were randomly allocated to receive propofol + saline (control group), propofol + sufentanil 0.1 µg/kg, propofol + dexmedetomidine 0.4 µg/kg, or propofol + ketamine 0.4 mg/kg. Mean arterial pressure, heart rate, pulse oximetry, pressure of end-tidal carbon dioxide, respiratory rate, and Ramsay sedation scale score were recorded. Induction time, procedure time, recovery time, propofol dose, and adverse events were also recorded. FINDINGS: During the sedation procedure, the AUC of HR was lowest in the propofol + dexmedetomidine group (all, P < 0.05), and the AUC of pulse oximetry was significantly higher in the propofol + dexmedetomidine and propofol + ketamine groups compared to the other 2 groups (both, P < 0.05). The propofol + dexmedetomidine group had the highest prevalences of hypotension and bradycardia, and the control group experienced the largest number of hypoxia episodes (all, P < 0.05). The control group consumed the highest dose of propofol, while the propofol + ketamine group needed the lowest dose (all, P < 0.05). IMPLICATIONS: The combination of propofol + ketamine 0.4 mg/kg maintained hemodynamic and respiratory stability, as evidenced by less hypotension, bradycardia, and hypoxia events, in elderly patients undergoing gastrointestinal endoscopy. China clinical trial registration (chictr.org.cn) ID: ChiCTR-INR-17013710.


Asunto(s)
Dexmedetomidina/uso terapéutico , Endoscopía Gastrointestinal , Hipnóticos y Sedantes/uso terapéutico , Ketamina/uso terapéutico , Propofol/uso terapéutico , Sufentanilo/uso terapéutico , Anciano , Anestesia , Dexmedetomidina/efectos adversos , Método Doble Ciego , Quimioterapia Combinada , Femenino , Humanos , Hipnóticos y Sedantes/efectos adversos , Ketamina/efectos adversos , Masculino , Propofol/efectos adversos , Sufentanilo/efectos adversos , Resultado del Tratamiento
17.
Biomed Res Int ; 2019: 2193706, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31205936

RESUMEN

The regulation of intracellular mitochondria degradation is mediated by mitophagy. While studies have shown that mitophagy can lead to mitochondrial dysfunction and cell damage, the role of Mdivi-1 and mitophagy remains unclear in acute lung injury (ALI) pathogenesis. In this study, we demonstrated that Mdivi-1, which is widely used as an inhibitor of mitophagy, ameliorated acute lung injury assessed by HE staining, pulmonary microvascular permeability assay, measurement of wet/dry weight (W/D) ratio, and oxygenation index (PaO2/FiO2) analysis. Then, the mitophagy related proteins were evaluated by western blot. The results indicated that LPS-induced activation of mitophagy was inhibited by Mdivi-1 treatment. In addition, we found that Mdivi-1 protected A549 cells against LPS-induced mitochondrial dysfunction. We also found that Mdivi-1 reduced pulmonary cell apoptosis in the LPS-challenged rats and protected pulmonary tissues from oxidative stress (represented by the content of superoxide dismutase, malondialdehyde and lipid peroxides in lung). Moreover, Mdivi-1 treatment ameliorated LPS-induced lung inflammatory response and cells recruitment. These findings indicate that Mdivi-1 mitigates LPS-induced apoptosis, oxidative stress, and inflammation in ALI, which may be associated with mitophagy inhibition. Thus, the inhibition of mitophagy may represent a potential therapy for treating ALI.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Mitocondrias/metabolismo , Mitofagia/efectos de los fármacos , Quinazolinonas/farmacología , Células A549 , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/patología , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Lipopolisacáridos/toxicidad , Masculino , Mitocondrias/patología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
18.
Lab Invest ; 99(6): 819-829, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30808930

RESUMEN

Mitophagy removes dysfunctional mitochondria and is known to play an important role in the pathogenesis of several diseases; however, the role of mitophagy in acute respiratory distress syndrome (ARDS) remains poorly understood. While we have previously demonstrated that polydatin (PD) improves lipopolysaccharide (LPS)-induced ARDS, the specific mechanism remains unclear. In present study, we aimed to determine whether PD activates Parkin-dependent mitophagy to protect against LPS-induced mitochondria-dependent apoptosis and lung injury. To establish the ARDS model, C57BL/6 mice were intratracheally injected with LPS (5 mg/kg) in vivo and Beas-2B cells were exposured to 0.5 mM LPS in vitro. Our results indicate that PD facilitates Parkin translocation to mitochondria and promotes mitophagy in ARDS-challenged mice and LPS-treated Beas-2B cells. However, PD-induced mitophagy was suppressed in Parkin-/- mice and Parkin siRNA transfected cells, indicating that PD activates Parkin-dependent mitophagy. Furthermore, the protective effects of PD against LPS-induced mitochondria-dependent apoptosis and lung injury were suppressed when Parkin was depleted both in vivo and in vitro. The inhibition of mitophagy with mitophagy inhibitor mitochondrial division inhibitor-1 in vivo and silencing of autophagy-related gene 7 in vitro also blocked the protective effects mediated by PD. Our data suggest that Parkin-dependent mitophagy induced by PD provides protection against mitochondria-dependent apoptosis in ARDS.


Asunto(s)
Apoptosis/efectos de los fármacos , Glucósidos/uso terapéutico , Mitofagia/efectos de los fármacos , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Estilbenos/uso terapéutico , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Evaluación Preclínica de Medicamentos , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Fallopia japonica , Glucósidos/farmacología , Masculino , Ratones Endogámicos C57BL , Fitoterapia , Síndrome de Dificultad Respiratoria/metabolismo , Estilbenos/farmacología
19.
Cell Death Dis ; 9(10): 932, 2018 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-30224699

RESUMEN

Parthanatos is a new form of programmed cell death. It has been recognized to be critical in cerebral ischemia-reperfusion injury, and reactive oxygen species (ROS) can induce parthanatos. Recent studies found that propofol, a widely used intravenous anesthetic agent, has an inhibitory effect on ROS and has neuroprotective in many neurological diseases. However, the functional roles and mechanisms of propofol in parthanatos remain unclear. Here, we discovered that the ROS-ER-calcium-mitochondria signal pathway mediated parthanatos and the significance of propofol in parthanatos. Next, we found that ROS overproduction would cause endoplasmic reticulum (ER) calcium release, leading to mitochondria depolarization with the loss of mitochondrial membrane potential. Mitochondria depolarization caused mitochondria to release more ROS, which, in turn, contributed to parthanatos. Also, we found that propofol inhibited parthanatos through impeding ROS overproduction, calcium release from ER, and mitochondrial depolarization in parthanatos. Importantly, our results indicated that propofol protected cerebral ischemia-reperfusion via parthanatos suppression, amelioration of mitochondria, and ER swelling. Our findings provide new insights into the mechanisms of how ER and mitochondria contribute to parthanatos. Furthermore, our studies elucidated that propofol has a vital role in parthanatos prevention in vivo and in vitro, and propofol can be a promising therapeutic approach for nerve injury patients.


Asunto(s)
Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , Propofol/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Western Blotting , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Línea Celular , Humanos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Transducción de Señal/efectos de los fármacos
20.
Gene ; 664: 90-100, 2018 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-29679758

RESUMEN

Propofol, a widely used intravenous anesthetic, was previously considered as a neuroprotective agent. Recently, however, accumulating evidence suggests that it may cause neurotoxicity, especially in the development of neural stem cells (NSCs). The potential mechanisms contributing to propofol-induced neurotoxicity during neurogenesis, such as those involving microRNAs (miRNAs), are still unknown. In this study, a total of 27 differentially expressed miRNAs were identified in our initial screen and 6 miRNAs were validated by qRT-PCR. Three miRNAs were up-regulated (miR-377-5p, miR-194-3p and miR-143-5p), and three were down-regulated (miR-3583-3p, miR-466b-5p and miR-410-5p). Following gene ontology and KEGG pathway enrichment analysis, Gabbr1, Canca1b and Gabbr2, which are enriched in the GABAergic synapse pathway, were selected as genes potentially playing a role in propofol-induced neurotoxicity. Gabbr1 and Cacna1b, which are targeted by miRNAs that are up-regulated following propofol exposure, showed decreased expression at the mRNA and protein levels. Gabbr2, targeted by miRNAs that were down-regulated following treatment with propofol, was up-regulated at both the levels of mRNA and protein expression. The two clusters of miRNAs that show differential expression following propofol exposure may act in a synergistic manner to regulate several genes simultaneously during the development of NSCs. Our results may contribute to clarify the molecular mechanism and provide potential therapeutic targets for propofol induced neurotoxicity.


Asunto(s)
Anestésicos Intravenosos/efectos adversos , Diferenciación Celular/efectos de los fármacos , MicroARNs/metabolismo , Células-Madre Neurales/fisiología , Propofol/efectos adversos , Animales , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo N/metabolismo , Biología Computacional , Femenino , Perfilación de la Expresión Génica , Células-Madre Neurales/efectos de los fármacos , Cultivo Primario de Células , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de GABA-B/genética , Receptores de GABA-B/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...