Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.173
Filtrar
1.
J Extracell Vesicles ; 13(9): e12495, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39254228

RESUMEN

Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) have shown anti-inflammatory potential in multiple inflammatory diseases. In the March 2022 issue of the Journal of Extracellular Vesicles, it was shown that EVs from human MSCs can suppress severe acute respiratory distress syndrome, coronavirus 2 (SARS-CoV-2) replication and can mitigate the production and release of infectious virions. We therefore hypothesized that MSC-EVs have an anti-viral effect in SARS-CoV-2 infection in vivo. We extended this question to ask whether also other respiratory viral infections could be treated by MSC-EVs. Adipose stem cell-derived EVs (ASC-EVs) were isolated using tangential flow filtration from conditioned media obtained from a multi-flask cell culture system. The effects of the ASC-EVs were tested  in Vero E6 cells in vitro. ASC-EVs were also given i.v. to SARS-CoV-2 infected Syrian Hamsters, and H1N1 influenza virus infected mice. The ASC-EVs attenuated SARS-CoV-2 virus replication in Vero E6 cells and reduced body weight and signs of lung injury in infected Syrian hamsters. Furthermore, ASC-EVs increased the survival rate of influenza A-infected mice and attenuated signs of lung injury. In summary, this study suggests that ASC-EVs can have beneficial therapeutic effects in models of virus-infection-associated acute lung injury and may potentially be developed to treat lung injury in humans.


Asunto(s)
Lesión Pulmonar Aguda , COVID-19 , Vesículas Extracelulares , Subtipo H1N1 del Virus de la Influenza A , Células Madre Mesenquimatosas , SARS-CoV-2 , Animales , Vesículas Extracelulares/metabolismo , Células Madre Mesenquimatosas/metabolismo , SARS-CoV-2/fisiología , COVID-19/terapia , Lesión Pulmonar Aguda/terapia , Lesión Pulmonar Aguda/virología , Ratones , Células Vero , Humanos , Chlorocebus aethiops , Infecciones por Orthomyxoviridae/terapia , Replicación Viral , Mesocricetus , Modelos Animales de Enfermedad , Masculino , Gripe Humana/terapia , Femenino
3.
Respir Res ; 25(1): 303, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39112999

RESUMEN

BACKGROUND: Acute lung injury (ALI) following pneumonia involves uncontrolled inflammation and tissue injury, leading to high mortality. We previously confirmed the significantly increased cargo content and extracellular vesicle (EV) production in thrombin-preconditioned human mesenchymal stromal cells (thMSCs) compared to those in naïve and other preconditioning methods. This study aimed to investigate the therapeutic efficacy of EVs derived from thMSCs in protecting against inflammation and tissue injury in an Escherichia coli (E. coli)-induced ALI mouse model. METHODS: In vitro, RAW 264.7 cells were stimulated with 0.1 µg/mL liposaccharides (LPS) for 1 h, then were treated with either PBS (LPS Ctrl) or 5 × 107 particles of thMSC-EVs (LPS + thMSC-EVs) for 24 h. Cells and media were harvested for flow cytometry and ELISA. In vivo, ICR mice were anesthetized, intubated, administered 2 × 107 CFU/100 µl of E. coli. 50 min after, mice were then either administered 50 µL saline (ECS) or 1 × 109 particles/50 µL of thMSC-EVs (EME). Three days later, the therapeutic efficacy of thMSC-EVs was assessed using extracted lung tissue, bronchoalveolar lavage fluid (BALF), and in vivo computed tomography scans. One-way analysis of variance with post-hoc TUKEY test was used to compare the experimental groups statistically. RESULTS: In vitro, IL-1ß, CCL-2, and MMP-9 levels were significantly lower in the LPS + thMSC-EVs group than in the LPS Ctrl group. The percentages of M1 macrophages in the normal control, LPS Ctrl, and LPS + thMSC-EV groups were 12.5, 98.4, and 65.9%, respectively. In vivo, the EME group exhibited significantly lower histological scores for alveolar congestion, hemorrhage, wall thickening, and leukocyte infiltration than the ECS group. The wet-dry ratio for the lungs was significantly lower in the EME group than in the ECS group. The BALF levels of CCL2, TNF-a, and IL-6 were significantly lower in the EME group than in the ECS group. In vivo CT analysis revealed a significantly lower percentage of damaged lungs in the EME group than in the ECS group. CONCLUSION: Intratracheal thMSC-EVs administration significantly reduced E. coli-induced inflammation and lung tissue damage. Overall, these results suggest therapeutically enhanced thMSC-EVs as a novel promising therapeutic option for ARDS/ALI.


Asunto(s)
Lesión Pulmonar Aguda , Vesículas Extracelulares , Células Madre Mesenquimatosas , Ratones Endogámicos ICR , Trombina , Animales , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/trasplante , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/microbiología , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/terapia , Ratones , Células Madre Mesenquimatosas/metabolismo , Células RAW 264.7 , Trombina/metabolismo , Escherichia coli , Masculino , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Infecciones por Escherichia coli/terapia , Resultado del Tratamiento , Modelos Animales de Enfermedad , Humanos
4.
ACS Nano ; 18(32): 21009-21023, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39087239

RESUMEN

Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), induce high morbidity and mortality rates, which challenge the present approaches for the treatment of ALI/ARDS. The clinically used photosensitizer verteporfin (VER) exhibits great potential in the treatment of acute lung injury and acute respiratory distress syndrome (ALI/ARDS) by regulating macrophage polarization and reducing inflammation. Nevertheless, its hydrophobic characteristics, nonspecificity, and constrained bioavailability hinder its therapeutic efficacy. In this work, we developed a type of VER-cored artificial exosome (EVM), which was produced by using mesoporous silica nanoparticles (MSNs) to load VER, followed by the exocytosis of internalized VER-MSNs from mouse bone marrow-derived mesenchymal stem cells (mBMSCs) without further modification. Both in vitro and in vivo assessments confirmed the powerful anti-inflammation induced by EVM. EVM also showed significant higher accumulation to inflammatory lungs compared with healthy ones, which was beneficial to the treatment of ALI/ARDS. EVM improved pulmonary function, attenuated lung injury, and reduced mortality in ALI mice with high levels of biocompatibility, exhibiting a 5-fold higher survival rate than the control. This type of artificial exosome emitted near-infrared light in the presence of laser activation, which endowed EVM with trackable ability both in vitro and in vivo. Our work developed a type of clinically used photosensitizer-loaded artificial exosome with membrane integrity and traceability. To the best of our knowledge, this kind of intracellularly synthesized artificial exosome was developed and showed great potential in ALI/ARDS therapy.


Asunto(s)
Lesión Pulmonar Aguda , Exosomas , Dióxido de Silicio , Animales , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/terapia , Ratones , Exosomas/metabolismo , Exosomas/química , Dióxido de Silicio/química , Verteporfina/farmacología , Verteporfina/química , Verteporfina/uso terapéutico , Nanopartículas/química , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Masculino , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Porosidad
7.
Front Biosci (Landmark Ed) ; 29(6): 217, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38940047

RESUMEN

BACKGROUND: Although umbilical cord mesenchymal stem cell (UCMSC) infusion has been proposed as a promising strategy for the treatment of acute lung injury (ALI), the parameters of UCMSC transplantation, such as infusion routes and doses, need to be further optimized. METHODS: In this study, we compared the therapeutic effects of UCMSCs transplanted via intravenous injection and intratracheal instillation on lipopolysaccharide-induced ALI using a rat model. Following transplantation, levels of inflammatory factors in serum; neutrophils, total white blood cells, and lymphocytes in bronchoalveolar lavage fluid (BALF); and lung damage levels were analyzed. RESULTS: The results indicated that UCMSCs administered via both intravenous and intratracheal routes were effective in alleviating ALI, as determined by analyses of arterial blood gas, lung histopathology, BALF contents, and levels of inflammatory factors. Comparatively, the intratracheal instillation of UCMSCs was found to result in lower levels of lymphocytes and total proteins in BALF, whereas greater reductions in the serum levels of tumor necrosis factor α (TNF-α) and interleukin 1ß (IL-1ß) were detected in rats receiving intravenously injected stem cells. CONCLUSIONS: Our findings in this study provide convincing evidence to indicate the efficacy of UCMSC therapy in the treatment of ALI mediated via different delivery routes, thereby providing a reliable theoretical basis for further clinical studies. Moreover, these findings imply that the effects obtained using the two assessed delivery routes for UCMSC transplantation are mediated via different mechanisms, which could be attributable to different cellular or molecular targets.


Asunto(s)
Lesión Pulmonar Aguda , Lipopolisacáridos , Trasplante de Células Madre Mesenquimatosas , Ratas Sprague-Dawley , Cordón Umbilical , Animales , Lesión Pulmonar Aguda/terapia , Lesión Pulmonar Aguda/inducido químicamente , Trasplante de Células Madre Mesenquimatosas/métodos , Cordón Umbilical/citología , Ratas , Masculino , Líquido del Lavado Bronquioalveolar/citología , Células Madre Mesenquimatosas/citología , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/metabolismo , Inyecciones Intravenosas
8.
Mol Biol Rep ; 51(1): 750, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38874700

RESUMEN

BACKGROUND: Acute lung injury (ALI) is a clinical syndrome characterized by pulmonary inflammation. Ultrashort wave diathermy (USWD) has been shown to be effective at in inhibiting ALI inflammation, although the underlying mechanism remains unclear. Previous studies have demonstrated that USWD generates a therapeutic thermal environment that aligns with the temperature required for heat shock protein 70 (HSP70), an endogenous protective substance. In this study, we examined the correlation between HSP70 and USWD in alleviating lung inflammation in ALI. METHODS: Forty-eight male C57BL/6 mice were randomly divided into control, model, USWD intervention (LU) 1, 2, and 3, and USWD preintervention (UL) 1, 2, and 3 groups (n = 6 in each group). The mice were pretreated with LPS to induce ALI. The UL1, 2, and 3 groups received USWD treatment before LPS infusion, while the LU1, 2, and 3 groups received USWD treatment after LPS infusion. Lung function and structure, inflammatory factor levels and HSP70 protein expression levels were detected. RESULTS: USWD effectively improved lung structure and function, and significantly reduced IL-1ß, IL-10, TGF-ß1, and TNF-α levels in both the USWD preintervention and intervention groups. However, HSP70 expression did not significantly differ across the experimental groups although the expression of TLR4 was significantly decreased, suggesting that USWD may have anti-inflammatory effects through multiple signaling pathways or that the experimental conditions should be restricted. CONCLUSIONS: Both USWD intervention and preintervention effectively reduced the inflammatory response, alleviated lung injury symptoms, and played a protective role in LPS-pretreated ALI mice. HSP70 was potentially regulated by USWD in this process, but further studies are urgently needed to elucidate the correlation and mechanism.


Asunto(s)
Lesión Pulmonar Aguda , Diatermia , Modelos Animales de Enfermedad , Proteínas HSP70 de Choque Térmico , Ratones Endogámicos C57BL , Neumonía , Animales , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/terapia , Proteínas HSP70 de Choque Térmico/metabolismo , Ratones , Masculino , Proyectos Piloto , Diatermia/métodos , Neumonía/metabolismo , Pulmón/metabolismo , Pulmón/patología , Lipopolisacáridos , Citocinas/metabolismo
9.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 36(5): 514-519, 2024 May.
Artículo en Chino | MEDLINE | ID: mdl-38845499

RESUMEN

OBJECTIVE: To investigate the effect of mild hypothermia on macrophage polarization in lipopolysaccharide (LPS)-induced acute lung injury (ALI) mice and to clarify its role in lung injury. METHODS: According to a random number table method, 18 male C57BL/6 mice were divided into sham operation group (Sham group), ALI normothermic model group (NT group) and ALI mild hypothermia treatment group (HT group), with 6 mice in each group. The ALI model in mice was established by the method of tracheal instillation of LPS, and temperature control was administered at 1 hour after surgery. The anus temperature in NT group was kept at 36-38?centigrade, while the anus temperature in HT group was kept at 32-34?centigrade. The target anus temperature in both groups were maintained for 6 hours and then slowly rewarmed to 36-38 centigrade. The Sham group was infused with an equal amount of physiological saline through the trachea without temperature control. After 24 hours of modeling, serum was collected and mice were sacrificed to obtain lung tissue. Pathological changes in lung tissue were observed under light microscopy and semi-quantitative lung injury score was performed. Enzyme linked immunosorbent assay (ELISA) was used to detect the serum levels of interleukins (IL-1ß, IL-10). Real-time quantitative polymerase chain reaction (RT-qPCR) was used to test the indicators of macrophage polarization, such as the mRNA expressions of CD86, IL-6, CD206 and arginase 1 (Arg1) in the lung tissue. The protein expression of M1 macrophage marker inducible nitric oxide synthase (iNOS) and M2 macrophage marker Arg1 were detected by Western blotting. RESULTS: Compared with the Sham group, the NT group appeared significant pulmonary hemorrhage and edema, thickened lung septum, inflammatory cell infiltration, and lung injury score was significantly increased; serum IL-1ß level was significantly elevated; IL-10 level was increased without statistical significance; the expressions of CD86 mRNA, IL-6 mRNA and iNOS protein were significantly elevated, and CD206 mRNA was significantly decreased; the mRNA and protein expressions of Arg1 decreased, but there were no significant differences. Compared with the NT group, the pathological injury of lung tissue in HT group was significantly reduced, and the lung injury score was significantly decreased (4.78±0.96 vs. 8.56±1.98, P < 0.01); serum IL-1ß level was decreased (ng/L: 13.52±1.95 vs. 27.18±3.87, P < 0.01), and IL-10 level was significantly increased (ng/L: 42.59±15.79 vs. 14.62±4.47, P < 0.01); IL-6 mRNA expression was decreased in lung tissue (2-ΔΔCt: 3.37±0.92 vs. 10.04±0.91, P < 0.05), the expression of M1 macrophage markers CD86 mRNA and iNOS protein were significantly decreased [CD86 mRNA (2-ΔΔCt): 0.52±0.16 vs. 1.95±0.33, iNOS protein (iNOS/ß-actin): 0.57±0.19 vs. 1.11±0.27, both P < 0.05], the expression of M2 macrophage markers CD206 mRNA, Arg1 mRNA and Arg1 protein were significantly increased [CD206 mRNA (2-ΔΔCt): 3.99±0.17 vs. 0.34±0.17, Arg1 mRNA (2-ΔΔCt): 2.33±0.73 vs. 0.94±0.23, Arg1 protein (Arg1/ß-actin): 0.96±0.09 vs. 0.31±0.11, all P < 0.05]. CONCLUSIONS: Mild hypothermia can alleviate the inflammatory response and protect lung tissue in ALI mice, which may be related to the inhibition of M1 macrophage polarization and promotion of M2 macrophage polarization.


Asunto(s)
Lesión Pulmonar Aguda , Lipopolisacáridos , Macrófagos , Ratones Endogámicos C57BL , Animales , Lesión Pulmonar Aguda/terapia , Masculino , Ratones , Macrófagos/metabolismo , Lipopolisacáridos/efectos adversos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Hipotermia Inducida , Interleucina-1beta/metabolismo , Modelos Animales de Enfermedad
10.
Transl Res ; 272: 111-125, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38897427

RESUMEN

Mammalian lung is the important organ for ventilation and exchange of air and blood. Fresh air and venous blood are constantly delivered through the airway and vascular tree to the alveolus. Based on this, the airways and alveolis are persistently exposed to the external environment and are easily suffered from toxins, irritants and pathogens. For example, acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a common cause of respiratory failure in critical patients, whose typical pathological characters are diffuse epithelial and endothelial damage resulting in excessive accumulation of inflammatory fluid in the alveolar cavity. The supportive treatment is the main current treatment for ALI/ARDS with the lack of targeted effective treatment strategies. However, ALI/ARDS needs more targeted treatment measures. Therefore, it is extremely urgent to understand the cellular and molecular mechanisms that maintain alveolar epithelial barrier and airway integrity. Previous researches have shown that the lung epithelial cells with tissue stem cell function have the ability to repair and regenerate after injury. Also, it is able to regulate the phenotype and function of innate immune cells involving in regeneration of tissue repair. Meanwhile, we emphasize that interaction between the lung epithelial cells and innate immune cells is more supportive to repair and regenerate in the lung epithelium following acute lung injury. We reviewed the recent advances in injury and repair of lung epithelial stem cells and innate immune cells in ALI/ARDS, concentrating on alveolar type 2 cells and alveolar macrophages and their contribution to post-injury repair behavior of ALI/ARDS through the latest potential molecular communication mechanisms. This will help to develop new research strategies and therapeutic targets for ALI/ARDS.


Asunto(s)
Lesión Pulmonar Aguda , Inmunidad Innata , Pulmón , Regeneración , Síndrome de Dificultad Respiratoria , Células Madre , Humanos , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/terapia , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/terapia , Pulmón/inmunología , Pulmón/patología , Animales , Células Epiteliales
11.
Am J Med Sci ; 368(3): 242-252, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38795966

RESUMEN

BACKGROUND: Alveolar capillary endothelial cell (EC) injury has a pivotal role in driving acute respiratory distress syndrome (ARDS) progression and maintaining endothelial homeostasis. A previous ex vivo study revealed that overexpression of homeobox B4 (HOXB4) in bone marrow mesenchymal stem cells (BMSCs) enhanced protection against lipopolysaccharide (LPS)-induced EC injury by activating the Wnt/ß-catenin pathway. This in vivo study was performed to verify whether BMSCs overexpressing HOXB4 exert similar protective effects on LPS-induced acute lung injury (ALI) in an animal model. METHODS: The ALI rat model was established by intraperitoneal injection of LPS. Wildtype BMSCs or BMSCs overexpressing HOXB4 were then injected via the tail vein. The lung characteristics of rats were visualized by computed tomography. Lung histopathological characteristics and collagen deposition were assessed by hematoxylin-eosin and Masson's staining, respectively, which were combined with the lung wet/dry ratio and proinflammatory factor levels in bronchoalveolar lavage fluid to further evaluate therapeutic effects. Expression of ß-catenin and VE-cadherin was assessed by western blotting and immunofluorescence. RESULTS: Compared with wildtype BMSCs, overexpression of HOXB4 optimized the therapeutic effects of BMSCs, which manifested as improvements in lung exudation and histopathological features, reduced lung collagen deposition, amelioration of lung permeability, attenuation of lung inflammation, and enhanced expression of ß-catenin and VE-cadherin proteins. CONCLUSIONS: HOXB4-overexpressing BMSCs optimized the protective effect against LPS-induced ALI by partially activating Wnt/ß-catenin signaling.


Asunto(s)
Lesión Pulmonar Aguda , Proteínas de Homeodominio , Lipopolisacáridos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Ratas Sprague-Dawley , Animales , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/terapia , Lesión Pulmonar Aguda/metabolismo , Ratas , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Masculino , Lipopolisacáridos/toxicidad , Modelos Animales de Enfermedad , Vía de Señalización Wnt , Endotoxinas/toxicidad
12.
PLoS One ; 19(5): e0303282, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38758742

RESUMEN

BACKGROUND: Severe acute lung failure (ALF) often necessitates veno-venous extracorporeal membrane oxygenation (VV-ECMO), where identifying predictors of weaning success and mortality remains crucial yet challenging. The study aims to identify predictors of weaning success and mortality in adults undergoing VV-ECMO for severe ALF, a gap in current clinical knowledge. METHODS AND ANALYSIS: PubMed, EMBASE, and the Cochrane Central Register of Controlled Trials will be searched for cohort studies examining the predictive factors of successful weaning and mortality in adult patients on VV-ECMO due to severe ALF. Risk of bias assessment will be conducted using the Newcastle-Ottawa scale for each included study. The primary outcomes will be successful weaning from VV-ECMO and all-cause mortality. Between-study heterogeneity will be evaluated using the I2 statistic. Sensitivity, subgroup, and meta-regression analyses will be performed to ascertain potential sources of heterogeneity and assess the robustness of our results. We will use the Grading of Recommendations, Assessment, Development, and Evaluations (GRADE) tool to recommend the level of evidence. DISCUSSION: This study seeks to provide clinically significant insights into predictors for weaning and mortality during VV-ECMO treatment for ALF, aiming to support clinical decisions and potentially influence health policy, thereby improving patient outcomes. ETHICS AND DISSEMINATION: Given the absence of direct engagement with human subjects or access to personal medical records, ethical approval for this study is deemed unnecessary. The study findings will be shared at a scientific conference either at the global or national level. Alternatively, the results will be presented for publication in a rigorously peer-reviewed journal regarding critical care medicine.


Asunto(s)
Lesión Pulmonar Aguda , Oxigenación por Membrana Extracorpórea , Humanos , Oxigenación por Membrana Extracorpórea/métodos , Adulto , Lesión Pulmonar Aguda/terapia , Lesión Pulmonar Aguda/mortalidad , Estudios de Cohortes
13.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 36(4): 430-434, 2024 Apr.
Artículo en Chino | MEDLINE | ID: mdl-38813641

RESUMEN

Hyperoxia-induced acute lung injury (HALI) is an important complication of clinical oxygen therapy, which is mainly characterized by acute respiratory distress syndrome (ARDS) in adults and broncho-pulmonary dysplasia (BPD) in infants. HALI seriously affects the prognosis and quality of life of patients, so it has received more and more attention. However, the pathogenesis of HALI is complex and unclear, and there is no clear treatment method at present. Non-coding RNA (ncRNA) is an important type of functional RNA transcriptome. Due to the lack of effective open reading frame, ncRNA does not have the function of coding proteins. However, ncRNA can still regulate gene expression at multiple levels and affect the occurrence and development of many diseases. In recent years, a large number of in vitro and in vivo studies have shown that ncRNA is involved in the pathogenesis of HALI and is of great significance. This article reviews the expression and significance of ncRNA in HALI, in order to provide new diagnosis and treatment ideas for the prevention and treatment of HALI.


Asunto(s)
Lesión Pulmonar Aguda , Hiperoxia , ARN no Traducido , Humanos , Hiperoxia/complicaciones , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/terapia , ARN no Traducido/genética , Animales
14.
Shock ; 62(2): 294-303, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38813932

RESUMEN

ABSTRACT: Introduction: We hypothesized extracellular vesicles (EVs) from preconditioned human-induced pluripotent stem cell-derived mesenchymal stem cells (iMSCs) attenuate LPS-induced acute lung injury (ALI) and endotoxemia. Methods: iMSCs were incubated with cell stimulation cocktail (CSC) and EVs were isolated. iMSC-EVs were characterized by size and EV markers. Biodistribution of intratracheal (IT), intravenous, and intraperitoneal injection of iMSC-EVs in mice was examined using IVIS. Uptake of iMSC-EVs in lung tissue, alveolar macrophages, and RAW264.7 cells was also assessed. C57BL/6 mice were treated with IT/IP iMSC-EVs or vehicle ± IT/IP LPS to induce ALI/acute respiratory distress syndrome and endotoxemia. Lung tissues, plasma, and bronchoalveolar lavage fluid (BALF) were harvested at 24 h. Lung histology, BALF neutrophil/macrophage, cytokine levels, and total protein concentration were measured to assess ALI and inflammation. Survival studies were performed using IP LPS in mice for 3 days. Results: iMSC-EV route of administration resulted in differential tissue distribution. iMSC-EVs were taken up by alveolar macrophages in mouse lung and cultured RAW264.7 cells. IT LPS-treated mice demonstrated marked histologic ALI, increased BALF neutrophils/macrophages and protein, and increased BALF and plasma TNF-α/IL-6 levels. These parameters were attenuated by 2 h before or 2 h after treatment with IT iMSC-EVs in ALI mice. Interestingly, the IT LPS-induced increase in IL-10 was augmented by iMSC-EVs. Mice treated with IP LPS showed increases in TNF-α and IL-6 that were downregulated by iMSC-EVs and LPS-induced mortality was ameliorated by iMSC-EVs. Administration of IT iMSC-EVs 2 h after LPS downregulated the increase in proinflammatory cytokines (TNF-α/IL-6) by LPS and further increased IL-10 levels. Conclusions: iMSC-EVs attenuate the inflammatory effects of LPS on cytokine levels in ALI and IP LPS in mice. LPS-induced mortality was improved with administration of iMSC-EVs.


Asunto(s)
Lesión Pulmonar Aguda , Endotoxemia , Vesículas Extracelulares , Células Madre Pluripotentes Inducidas , Lipopolisacáridos , Células Madre Mesenquimatosas , Ratones Endogámicos C57BL , Animales , Vesículas Extracelulares/metabolismo , Ratones , Lipopolisacáridos/toxicidad , Endotoxemia/terapia , Endotoxemia/inducido químicamente , Células Madre Mesenquimatosas/metabolismo , Lesión Pulmonar Aguda/terapia , Lesión Pulmonar Aguda/inducido químicamente , Células RAW 264.7 , Masculino , Humanos
15.
Cells ; 13(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38727303

RESUMEN

Small interfering RNA (siRNA) holds significant therapeutic potential by silencing target genes through RNA interference. Current clinical applications of siRNA have been primarily limited to liver diseases, while achievements in delivery methods are expanding their applications to various organs, including the lungs. Cholesterol-conjugated siRNA emerges as a promising delivery approach due to its low toxicity and high efficiency. This study focuses on developing a cholesterol-conjugated anti-Il6 siRNA and the evaluation of its potency for the potential treatment of inflammatory diseases using the example of acute lung injury (ALI). The biological activities of different Il6-targeted siRNAs containing chemical modifications were evaluated in J774 cells in vitro. The lead cholesterol-conjugated anti-Il6 siRNA after intranasal instillation demonstrated dose-dependent therapeutic effects in a mouse model of ALI induced by lipopolysaccharide (LPS). The treatment significantly reduced Il6 mRNA levels, inflammatory cell infiltration, and the severity of lung inflammation. IL6 silencing by cholesterol-conjugated siRNA proves to be a promising strategy for treating inflammatory diseases, with potential applications beyond the lungs.


Asunto(s)
Lesión Pulmonar Aguda , Colesterol , Interleucina-6 , ARN Interferente Pequeño , Animales , Ratones , Lesión Pulmonar Aguda/terapia , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/metabolismo , Línea Celular , Colesterol/química , Colesterol/farmacología , Modelos Animales de Enfermedad , Interleucina-6/metabolismo , Interleucina-6/genética , Lipopolisacáridos , Pulmón/patología , Pulmón/metabolismo , Ratones Endogámicos C57BL , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología
16.
Cell Commun Signal ; 22(1): 293, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802896

RESUMEN

BACKGROUND: Acute respiratory distress syndrome (ARDS) is a severe and fatal disease. Although mesenchymal stem cell (MSC)-based therapy has shown remarkable efficacy in treating ARDS in animal experiments, clinical outcomes have been unsatisfactory, which may be attributed to the influence of the lung microenvironment during MSC administration. Extracellular vesicles (EVs) derived from endothelial cells (EC-EVs) are important components of the lung microenvironment and play a crucial role in ARDS. However, the effect of EC-EVs on MSC therapy is still unclear. In this study, we established lipopolysaccharide (LPS) - induced acute lung injury model to evaluate the impact of EC-EVs on the reparative effects of bone marrow-derived MSC (BM-MSC) transplantation on lung injury and to unravel the underlying mechanisms. METHODS: EVs were isolated from bronchoalveolar lavage fluid of mice with LPS - induced acute lung injury and patients with ARDS using ultracentrifugation. and the changes of EC-EVs were analysed using nanoflow cytometry analysis. In vitro assays were performed to establish the impact of EC-EVs on MSC functions, including cell viability and migration, while in vivo studies were performed to validate the therapeutic effect of EC-EVs on MSCs. RNA-Seq analysis, small interfering RNA (siRNA), and a recombinant lentivirus were used to investigate the underlying mechanisms. RESULTS: Compared with that in non-ARDS patients, the quantity of EC-EVs in the lung microenvironment was significantly greater in patients with ARDS. EVs derived from lipopolysaccharide-stimulated endothelial cells (LPS-EVs) significantly decreased the viability and migration of BM-MSCs. Furthermore, engrafting BM-MSCs pretreated with LPS-EVs promoted the release of inflammatory cytokines and increased pulmonary microvascular permeability, aggravating lung injury. Mechanistically, LPS-EVs reduced the expression level of isocitrate dehydrogenase 2 (IDH2), which catalyses the formation of α-ketoglutarate (α-KG), an intermediate product of the tricarboxylic acid (TCA) cycle, in BM-MSCs. α-KG is a cofactor for ten-eleven translocation (TET) enzymes, which catalyse DNA hydroxymethylation in BM-MSCs. CONCLUSIONS: This study revealed that EC-EVs in the lung microenvironment during ARDS can affect the therapeutic efficacy of BM-MSCs through the IDH2/TET pathway, providing potential strategies for improving the therapeutic efficacy of MSC-based therapy in the clinic.


Asunto(s)
Células Endoteliales , Vesículas Extracelulares , Isocitrato Deshidrogenasa , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Síndrome de Dificultad Respiratoria , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/trasplante , Animales , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Síndrome de Dificultad Respiratoria/terapia , Síndrome de Dificultad Respiratoria/metabolismo , Células Endoteliales/metabolismo , Humanos , Ratones , Trasplante de Células Madre Mesenquimatosas/métodos , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Ratones Endogámicos C57BL , Masculino , Lipopolisacáridos/farmacología , Transducción de Señal , Lesión Pulmonar Aguda/terapia , Lesión Pulmonar Aguda/metabolismo , Movimiento Celular
17.
Stem Cell Res Ther ; 15(1): 95, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38566259

RESUMEN

BACKGROUND: Human adipose stromal cells-derived extracellular vesicles (haMSC-EVs) have been shown to alleviate inflammation in acute lung injury (ALI) animal models. However, there are few systemic studies on clinical-grade haMSC-EVs. Our study aimed to investigate the manufacturing, quality control (QC) and preclinical safety of clinical-grade haMSC-EVs. METHODS: haMSC-EVs were isolated from the conditioned medium of human adipose MSCs incubated in 2D containers. Purification was performed by PEG precipitation and differential centrifugation. Characterizations were conducted by nanoparticle tracking analysis, transmission electron microscopy (TEM), Western blotting, nanoflow cytometry analysis, and the TNF-α inhibition ratio of macrophage [after stimulated by lipopolysaccharide (LPS)]. RNA-seq and proteomic analysis with liquid chromatography tandem mass spectrometry (LC-MS/MS) were used to inspect the lot-to-lot consistency of the EV products. Repeated toxicity was evaluated in rats after administration using trace liquid endotracheal nebulizers for 28 days, and respiratory toxicity was evaluated 24 h after the first administration. In vivo therapeutic effects were assessed in an LPS-induced ALI/ acute respiratory distress syndrome (ARDS) rat model. RESULTS: The quality criteria have been standardized. In a stability study, haMSC-EVs were found to remain stable after 6 months of storage at - 80°C, 3 months at - 20 °C, and 6 h at room temperature. The microRNA profile and proteome of haMSC-EVs demonstrated suitable lot-to-lot consistency, further suggesting the stability of the production processes. Intratracheally administered 1.5 × 108 particles/rat/day for four weeks elicited no significant toxicity in rats. In LPS-induced ALI/ARDS model rats, intratracheally administered haMSC-EVs alleviated lung injury, possibly by reducing the serum level of inflammatory factors. CONCLUSION: haMSC-EVs, as an off-shelf drug, have suitable stability and lot-to-lot consistency. Intratracheally administered haMSC-EVs demonstrated excellent safety at the tested dosages in systematic preclinical toxicity studies. Intratracheally administered haMSC-EVs improved the lung function and exerted anti-inflammatory effects on LPS-induced ALI/ARDS model rats.


Asunto(s)
Lesión Pulmonar Aguda , Vesículas Extracelulares , Células Madre Mesenquimatosas , Síndrome de Dificultad Respiratoria , Humanos , Ratas , Animales , Cromatografía Liquida , Proteómica , Lipopolisacáridos/farmacología , Espectrometría de Masas en Tándem , Lesión Pulmonar Aguda/terapia , Síndrome de Dificultad Respiratoria/terapia , Obesidad , Control de Calidad , Vesículas Extracelulares/fisiología , Células Madre Mesenquimatosas/fisiología
18.
Respir Care ; 69(8): 959-967, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-38653557

RESUMEN

BACKGROUND: In recent years, acute lung injury (ALI) and ARDS have emerged as critical health concerns, drawing considerable attention from clinicians. The volume of published articles on ALI/ARDS is on the rise, indicating the expanding research interest in this field. However, the precise quantity and quality of studies on ALI/ARDS remain unclear. Consequently, we employed bibliometric and visual techniques to comprehensively analyze the patterns and focal points of these articles. METHODS: To investigate the characteristics of highly referenced papers on ALI/ARDS and offer insights into the progress and advancements in research on ALI/ARDS, we conducted a comprehensive search in the core Web of Science database for cited articles using the terms "ALI," "acute lung injury," "ARDS," or "acute respiratory distress syndrome." A total of 60,282 citations were retrieved by narrowing the scope to reviews, articles, and publications in English. From the obtained citations, we selected materials for analysis from the top 100 articles with the highest number of citations. Subsequently, the articles were visualized and analyzed using VOSviewer, CiteSpace, and bibliometric techniques. This analysis focused on identifying trends related to authors, journals, countries, institutions, collaborative networks, key words, and other relevant factors in the field of ALI/ARDS research. RESULTS: Among the top 100 cited articles, the highest and lowest number of citations were 6,957 and 451, respectively. The 100 articles were published between 1991-2020, with a peak in publications observed in 2004, 2005, and 2012 (no. = 7). Among 29 journals, The New England Journal of Medicine (no. = 21) had the highest number of publications, followed by the American Journal of Respiratory and Critical Care Medicine (no. = 14). Among the 29 countries represented in the top 100 cited articles, the United States (no. = 51) emerged as the leading country in the number of publications, followed by Canada (no. = 19) (there was some overlap in paper output between countries due to co-publication). The 3 predominant keywords identified in studies within the ALI/ARDS domain were ALI, mechanical ventilation, and PEEP. CONCLUSIONS: This study provides a historical perspective on the scientific advancements in ALI/ARDS research, highlighting the need for further investigation and development in specific areas within the field. Bibliometric analyses revealed that the United States is the predominant force in the field of ALI/ARDS, contributing significantly to its development. Through an examination of highly cited papers on ALI/ARDS, we have identified global research trends, assessed the quality of studies, and identified hot topics in the field of ALI/ARDS.


Asunto(s)
Lesión Pulmonar Aguda , Bibliometría , Síndrome de Dificultad Respiratoria , Humanos , Síndrome de Dificultad Respiratoria/terapia , Lesión Pulmonar Aguda/terapia , Publicaciones Periódicas como Asunto/estadística & datos numéricos
19.
Biochem Biophys Res Commun ; 714: 149973, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38657444

RESUMEN

Acute respiratory distress syndrome (ARDS) is characterized by acute diffuse inflammatory lung injury with a high mortality rate. Mesenchymal stromal cells (MSC) are pluripotent adult cells that can be extracted from a variety of tissues, including the lung. Lung-resident MSC (LR-MSC) located around vascular vessels and act as important regulators of lung homeostasis, regulating the balance between lung injury and repair processes. LR-MSC support the integrity of lung tissue by modulating immune responses and releasing trophic factors. Studies have reported that the STING pathway is involved in the progression of lung injury inflammation, but the specific mechanism is unclear. In this study, we found that STING deficiency could ameliorate lipopolysaccharides (LPS)-induced acute lung injury, STING knockout (STING KO) LR-MSC had an enhanced treatment effect on acute lung injury. STING depletion protected LR-MSC from LPS-induced apoptosis. RNA-sequencing and Western blot results showed that STING KO LR-MSC expressed higher levels of MSC immunoregulatory molecules, such as Igfbp4, Icam1, Hgf and Cox2, than WT LR-MSC. This study highlights that LR-MSC have a therapeutic role in acute lung injury, and we demonstrate that STING deficiency can enhance the immunomodulatory function of LR-MSC in controlling lung inflammation. Thus, STING can be used as an intervention target to enhance the therapeutic effect of MSC.


Asunto(s)
Lesión Pulmonar Aguda , Lipopolisacáridos , Pulmón , Proteínas de la Membrana , Células Madre Mesenquimatosas , Ratones Endogámicos C57BL , Animales , Lipopolisacáridos/toxicidad , Células Madre Mesenquimatosas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/deficiencia , Pulmón/patología , Pulmón/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/terapia , Lesión Pulmonar Aguda/metabolismo , Ratones , Ratones Noqueados , Apoptosis , Masculino
20.
Acta Biomater ; 181: 308-316, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38570107

RESUMEN

Acute lung injury (ALI) represents a critical respiratory condition typified by rapid-onset lung inflammation, contributing to elevated morbidity and mortality rates. Central to ALI pathogenesis lies macrophage dysfunction, characterized by an overabundance of pro-inflammatory cytokines and a shift in metabolic activity towards glycolysis. This study emphasizes the crucial function of glucose metabolism in immune cell function under inflammatory conditions and identifies hexokinase 2 (HK2) as a key regulator of macrophage metabolism and inflammation. Given the limitations of HK2 inhibitors, we propose the CRISPR/Cas9 system for precise HK2 downregulation. We developed an aerosolized core-shell liposomal nanoplatform (CSNs) complexed with CaP for efficient drug loading, targeting lung macrophages. Various CSNs were synthesized to encapsulate an mRNA based CRISPR/Cas9 system (mCas9/gHK2), and their gene editing efficiency and HK2 knockout were examined at both gene and protein levels in vitro and in vivo. The CSN-mCas9/gHK2 treatment demonstrated a significant reduction in glycolysis and inflammation in macrophages. In an LPS-induced ALI mouse model, inhaled CSN-mCas9/gHK2 mitigated the proinflammatory tumor microenvironment and reprogrammed glucose metabolism in the lung, suggesting a promising strategy for ALI prevention and treatment. This study highlights the potential of combining CRISPR/Cas9 gene editing with inhalation delivery systems for effective, localized pulmonary disease treatment, underscoring the importance of targeted gene modulation and metabolic reprogramming in managing ALI. STATEMENT OF SIGNIFICANCE: This study investigates an inhalable CRISPR/Cas9 gene editing system targeting pulmonary macrophages, with the aim of modulating glucose metabolism to alleviate Acute Lung Injury (ALI). The research highlights the role of immune cell metabolism in inflammation, as evidenced by changes in macrophage glucose metabolism and a notable reduction in pulmonary edema and inflammation. Additionally, observed alterations in macrophage polarization and cytokine levels in bronchoalveolar lavage fluid suggest potential therapeutic implications. These findings not only offer insights into possible ALI treatments but also contribute to the understanding of immune cell metabolism in inflammatory diseases, which could be relevant for various inflammatory and metabolic disorders.


Asunto(s)
Lesión Pulmonar Aguda , Sistemas CRISPR-Cas , Hexoquinasa , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/terapia , Animales , Ratones , Hexoquinasa/genética , Hexoquinasa/metabolismo , Ratones Endogámicos C57BL , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Administración por Inhalación , Liposomas/química , Células RAW 264.7 , Masculino , Reprogramación Celular/efectos de los fármacos , Edición Génica , Glucólisis/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA