Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Stem Cells ; 41(5): 468-481, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-36827175

RESUMEN

Mesenchymal stem cells (MSCs) therapy could efficiently attenuate LPS-induced acute lung injury and Pseudomonas aeruginosa (PA)-induced acute pneumonia. However, the underlying molecular mechanisms are still elusive. Here, we report that PA-derived outer membrane vesicles (OMVs) trigger mouse primary adipose tissue-derived mesenchymal stem cells (ASCs) to upregulate cyclic GMP-AMP synthase (cGAS) for sensing of double-stranded DNA (dsDNA) and the expression of interleukin (IL)-7. Loss of cGAS-interferon (IFN)-ß axis abolished the protective function of ASCs to PA-induced acute pneumonia in mice. Mechanistically, OMVs-delivered PA dsDNA primes cGAS-stimulator of interferon genes (STING) signaling pathway and increases the IL-7 production in ASCs via IFN-ß signaling. Meanwhile, dsDNA-primed ASCs furthermore amplifies IL-7 expression in primary lung epithelial cells and mouse lung epithelial (MLE)-12 cell line via increased IFN-ß. Our findings thus implicate a molecular mechanism that ASCs recognize PA-OMVs-derived dsDNA to secrete IL-7 via activating cGAS, suggesting a potential therapeutic strategy of ASCs transfer for PA-induced lung infection and inflammation.


Asunto(s)
Interferón Tipo I , Neumonía , Ratones , Animales , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Interleucina-7 , Proteínas de la Membrana/genética , Interferón Tipo I/metabolismo , ADN/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Neumonía/terapia
2.
Small ; 18(38): e2203035, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35988138

RESUMEN

Capacity degradation and destructive hazards are two major challenges for the operation of lithium-ion batteries at high temperatures. Although adding flame retardants or fire extinguishing agents can provide one-off self-protection in case of emergency overheating, it is desirable to directly regulate battery operation according to the temperature. Herein, smart self-protecting aqueous lithium-ion batteries are developed using thermos-responsive separators prepared through in situ polymerization on the hydrophilic separator. The thermos-responsive separator blocks the lithium ion transport channels at high temperature and reopens when the battery cools down; more importantly, this transition is reversible. The influence of lithium salts on the thermos-responsive behaviors of the hydrogels is investigated. Then suitable lithium salt (LiNO3 ) and concentration (1 m) are selected in the electrolyte to achieve self-protection without sacrificing battery performance. The shut-off temperature can be tuned from 30 to 80 °C by adjusting the hydrophilic and hydrophobic moiety ratio in the hydrogel for targeted applications. This self-protecting LiMn2 O4 /carbon coated LiTi2 (PO4 )3 (LMO/C-LTP) battery shows promise for smart energy storage devices with high safety and extended lifespan in case of high operating temperatures.

3.
Curr Opin Infect Dis ; 34(2): 156-168, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33605620

RESUMEN

PURPOSE OF REVIEW: Although experimental evidence supports the use of nebulized antibiotics in ventilator-associated pneumonia (VAP), two recent multicenter randomized controlled trials (RCTs) have failed to demonstrate any benefit in VAP caused by Gram-negative bacteria (GNB). This review examines the methodological requirements concerning future RCTs. RECENT FINDINGS: High doses of nebulized antibiotics are required to reach the infected lung parenchyma. Breath-synchronized nebulizers do not allow delivery of high doses. Mesh nebulizers perform better than jet nebulizers. Epithelial lining fluid concentrations do not reflect interstitial lung concentrations in patients receiving nebulized antibiotics. Specific ventilator settings for optimizing lung deposition require sedation to avoid patient's asynchrony with the ventilator. SUMMARY: Future RCTs should compare a 3-5 day nebulization of amikacin or colistimethate sodium (CMS) to a 7-day intravenous administration of a new cephalosporine/ß-lactamase inhibitor. Inclusion criteria should be a VAP or ventilator-associated tracheobronchitis caused by documented extensive-drug or pandrug resistant GNB. If the GNB remains susceptible to aminoglycosides, nebulized amikacin should be administered at a dose of 40 mg/kg/day. If resistant to aminoglycosides, nebulized CMS should be administered at a dose of 15 millions international units (IU)/day. In VAP caused by pandrug-resistant GNB, 15 millions IU/day nebulized CMS (substitution therapy) should be compared with a 9 millions IU/day intravenous CMS.


Asunto(s)
Antibacterianos/administración & dosificación , Neumonía Asociada al Ventilador/tratamiento farmacológico , Administración por Inhalación , Antibacterianos/química , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/fisiología , Humanos , Nebulizadores y Vaporizadores , Neumonía Asociada al Ventilador/microbiología , Ensayos Clínicos Controlados Aleatorios como Asunto
4.
Clin Infect Dis ; 67(suppl_2): S146-S152, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30423049

RESUMEN

Background: The microbial etiology and mortality risk factors of ventilator-associated pneumonia (VAP) have not been investigated extensively in Shanghai. Methods: VAP cases were identified from the patients hospitalized during the period from 1 January 2013 to 30 December 2017 in Shanghai. The relevant data were reviewed and analyzed retrospectively. Results: One hundred ninety-four VAP cases were included in this analysis. The overall mortality rate was 32.47%. The respiratory pathogens isolated from these patients included 212 bacterial strains and 54 fungal strains. The leading pathogens were Acinetobacter baumannii (33.96%), Klebsiella pneumoniae (23.58%), Pseudomonas aeruginosa (19.81%), and Staphylococcus aureus (7.08%). Candida colonization was associated with higher mortality of VAP patients compared to those without Candida colonization (45.45% vs 28.67%, P < .05). The VAP patients with Candida colonization also showed higher prevalence of P. aeruginosa, carbapenem-resistant P. aeruginosa (CRPA), K. pneumoniae, carbapenem-resistant K. pneumoniae (CRKP), A. baumannii, and carbapenem-resistant A. baumannii (CRAB) (P < .05). VAP nonsurvivors had higher prevalence of CRPA, K. pneumoniae, CRAB, and Candida than VAP survivors (P < .05). Multivariate analysis showed that prior antibiotic use was a significant risk factor for Candida colonization, while hypertension and length of hospital stay were significant risk factors of VAP mortality (P < .05). Conclusions: The top pathogens of VAP patients in Shanghai tertiary teaching hospitals are A. baumannii, K. pneumoniae, and P. aeruginosa, with high prevalence of carbapenem resistance. Carbapenem-resistant bacterial pathogens and Candida may predict poor outcome.


Asunto(s)
Bacterias/aislamiento & purificación , Candida/aislamiento & purificación , Neumonía Asociada al Ventilador/microbiología , Neumonía Asociada al Ventilador/mortalidad , Anciano , Anciano de 80 o más Años , Antibacterianos/uso terapéutico , Bacterias/efectos de los fármacos , Candida/efectos de los fármacos , China/epidemiología , Farmacorresistencia Bacteriana Múltiple , Femenino , Hospitales de Enseñanza , Humanos , Hipertensión/complicaciones , Tiempo de Internación , Masculino , Persona de Mediana Edad , Neumonía Asociada al Ventilador/diagnóstico , Pronóstico , Estudios Retrospectivos , Factores de Riesgo
5.
Int J Cancer ; 142(4): 769-778, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29023689

RESUMEN

The functional role of respiratory microbiota has attracted an accumulating attention recently. However, the role of respiratory microbiome in lung carcinogenesis is mostly unknown. Our study aimed to characterize and compare bilateral lower airway microbiome of lung cancer patients with unilateral lobar masses and control subjects. Protected bronchial specimen brushing samples were collected from 24 lung cancer patients with unilateral lobar masses (paired samples from cancerous site and the contralateral noncancerous site) and 18 healthy controls undergoing bronchoscopies and further analyzed by 16S rRNA amplicon sequencing. As results, significant decreases in microbial diversity were observed in patients with lung cancer in comparison to the controls, alpha diversity steadily declined from healthy site to noncancerous to cancerous site. Genus Streptococcus was significantly more abundant in cancer cases than the controls, while Staphylococcus was more abundant in the controls. The area under the curve of genus Streptococcus used to predict lung cancer was 0.693 (sensitivity = 87.5%, specificity = 55.6%). The abundance of genus Streptococcus and Neisseria displayed an increasing trend whereas Staphylococcus and Dialister gradually declined from healthy to noncancerous to cancerous site. Collectively, lung cancer-associated microbiota profile is distinct from that found in healthy controls, and the altered cancer-associated microbiota is not restricted to tumor tissue. The genus Streptococcus was abundant in lung cancer patients and exhibited moderate classification potential. The gradual microbiota profile shift from healthy site to noncancerous to paired cancerous site suggested a change of the microenvironment associated with the development of lung cancer.


Asunto(s)
Neoplasias Pulmonares/microbiología , Microbiota , Adulto , Anciano , Broncoscopía , Estudios de Casos y Controles , Femenino , Humanos , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Neisseria/aislamiento & purificación , Infecciones por Neisseriaceae/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus/aislamiento & purificación
6.
Stem Cells ; 35(7): 1849-1859, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28376568

RESUMEN

Microvesicles (MVs) derived from human mesenchymal stem cells (MSC MVs) were demonstrated to ameliorate inflammation in lungs. We have found their content of mRNA for keratinocyte growth factor was partly involved in their therapeutic effects. As MSC MVs also contained a substantial quantity of angiopoietin-1 (Ang-1) mRNA, which plays an essential role in vascular stabilization and resolving inflammation, we hypothesized that Ang-1 mRNA might similarly account for a part of their therapeutic effects. We downregulated Ang-1 mRNA expression in MVs, using a lentivirus vector carrying Ang-1 short hairpin RNA to transfect MSCs. A mouse model of lipopolysaccharide induced acute lung injury (ALI) was used in vivo. We also studied in vitro interactions between Ang-1 mRNA deficient MVs on macrophages and human lung microvascular endothelial cells. Compared with negative control, Ang-1 mRNA deficient MVs increased the influx of neutrophils and macrophage inflammatory protein-2 levels in bronchoalveolar lavage fluid by 136% and 105%, respectively, suggesting a deteriorative lung inflammation and a failure to restore pulmonary capillary permeability assessed by Evan's blue dye and bronchoalveolar lavage albumin level. In vitro, the addition of Ang-1 mRNA deficient MVs failed to maintain the integrity of endotoxin-stimulated microvascular endothelial cells and abrogated the decrease in tumor necrosis factor-α level and the increase in interleukin-10 level mediated by negative control in RAW 264.7 cells. In summary, the therapeutic effects of MVs in ALI, and their immunomodulatory properties on macrophages were partly mediated through their content of Ang-1 mRNA. Stem Cells 2017;35:1849-1859.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Angiopoyetina 1/genética , Micropartículas Derivadas de Células/metabolismo , Pulmón/metabolismo , Células Madre Mesenquimatosas/metabolismo , ARN Mensajero/genética , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 1/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/química , Permeabilidad Capilar/genética , Micropartículas Derivadas de Células/química , Micropartículas Derivadas de Células/trasplante , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Humanos , Interleucina-10/genética , Interleucina-10/metabolismo , Lipopolisacáridos , Pulmón/patología , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/genética , Neutrófilos/metabolismo , Neutrófilos/patología , Cultivo Primario de Células , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
7.
Stem Cells ; 33(7): 2331-42, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25788456

RESUMEN

RATIONALE: New strategies for treating Pseudomonas aeruginosa pulmonary infection are urgently needed. Adipose tissue-derived mesenchymal stem cells (ASCs) may have a potential therapeutic role in P. aeruginosa-induced pulmonary infection. METHODS: The therapeutic and mechanistic effects of ASCs on P. aeruginosa pulmonary infection were evaluated in a murine model of P. aeruginosa pneumonia. RESULTS: ASCs exhibited protective effects against P. aeruginosa pulmonary infection, evidenced by reduced bacterial burdens, inhibition of alveolar neutrophil accumulation, decreased levels of myeloperoxidase, macrophage inflammatory protein-2 and total proteins in broncho-alveolar lavage fluid (BALF), and attenuated severity of lung injury. ASCs had no effects on BALF and serum levels of keratinocyte growth factor or Ang-1. ASCs had no effects on the levels of insulin growth factor 1 (IGF-1) in BALF, but increased IGF-1 levels in serum. ASCs inhibited the overproduction of prostaglandin E2 (PGE2 ) by decreasing the expression of cyclooxygenase-2 (COX2) and enhancing the expression of 15-PGDH. In addition, the addition of exogenous PGE2 with ASCs abolished many of the protective effects of ASCs, and administrating PGE2 alone exacerbated lung infection. By inhibiting production of PGE2 , ASCs improved phagocytosis and the bactericidal properties of macrophages. Furthermore suppressing PGE2 signaling by COX2 inhibition or EP2 inhibition exhibited protective effects against pulmonary infection as well. CONCLUSIONS: In a murine model of P. aeruginosa pneumonia, ASCs exhibited protective effects by inhibiting production of PGE2 , which subsequently improved phagocytosis and the bactericidal properties of macrophages. ASCs may provide a new strategy for managing pulmonary infection caused by P. aeruginosa.


Asunto(s)
Tejido Adiposo/metabolismo , Dinoprostona/metabolismo , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/metabolismo , Pseudomonas aeruginosa/patogenicidad , Animales , Modelos Animales de Enfermedad , Enfermedades Pulmonares/patología , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL
8.
Am J Respir Crit Care Med ; 192(3): 324-36, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26067592

RESUMEN

RATIONALE: Microvesicles (MVs) are anuclear fragments of cells released from the endosomal compartment or shed from surface membranes. We and other investigators demonstrated that MVs released by mesenchymal stem cells (MSCs) were as effective as the cells themselves in inflammatory injuries, such as after endotoxin-induced acute lung injury. However, the therapeutic effects of MVs in an infectious model of acute lung injury remain unknown. OBJECTIVES: We investigated the effects of human MSC MVs on lung inflammation, protein permeability, bacterial clearance, and survival after severe bacterial pneumonia. METHODS: We tested the effects of MVs derived from human MSCs on Escherichia coli pneumonia in mice. We also studied the interactions between MVs and human monocytes and human alveolar epithelial type 2 cells. MEASUREMENTS AND MAIN RESULTS: Administration of MVs derived from human MSCs improved survival in part through keratinocyte growth factor secretion and decreased the influx of inflammatory cells, cytokines, protein, and bacteria in mice injured with bacterial pneumonia. In primary cultures of human monocytes or alveolar type 2 cells, the uptake of MVs was mediated by CD44 receptors, which were essential for the therapeutic effects. MVs enhanced monocyte phagocytosis of bacteria while decreasing inflammatory cytokine secretion and increased intracellular ATP levels in injured alveolar epithelial type 2 cells. Prestimulation of MSCs with a toll-like receptor 3 agonist further enhanced the therapeutic effects of the released MVs. CONCLUSIONS: MVs derived from human MSCs were as effective as the parent stem cells in severe bacterial pneumonia.


Asunto(s)
Lesión Pulmonar Aguda/terapia , Micropartículas Derivadas de Células/trasplante , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas , Neumonía Bacteriana/terapia , Lesión Pulmonar Aguda/microbiología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
9.
Stem Cells ; 32(1): 116-25, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23939814

RESUMEN

We previously found that human mesenchymal stem cells (MSC) or its conditioned medium restored lung protein permeability and reduced alveolar inflammation following Escherichia coli endotoxin-induced acute lung injury (ALI) in an ex vivo perfused human lung in part through the secretion of soluble factors such as keratinocyte growth factor (KGF). Recently, MSC were found to release microvesicles (MVs) that were biologically active because of the presence of mRNA or miRNA with reparative properties. MVs are circular fragments of membrane released from the endosomal compartment as exosomes or shed from the surface membranes. These studies were designed to determine if MVs released by human bone marrow derived MSCs would be effective in restoring lung protein permeability and reducing inflammation in E. coli endotoxin-induced ALI in C57BL/6 mice. The intratracheal instillation of MVs improved several indices of ALI at 48 hours. Compared to endotoxin-injured mice, MVs reduced extravascular lung water by 43% and reduced total protein levels in the bronchoalveolar lavage (BAL) fluid by 35%, demonstrating a reduction in pulmonary edema and lung protein permeability. MVs also reduced the influx of neutrophils and macrophage inflammatory protein-2 levels in the BAL fluid by 73% and 49%, respectively, demonstrating a reduction in inflammation. KGF siRNA-pretreatment of MSC partially eliminated the therapeutic effects of MVs released by MSCs, suggesting that KGF protein expression was important for the underlying mechanism. In summary, human MSC-derived MVs were therapeutically effective following E. coli endotoxin-induced ALI in mice in part through the expression of KGF mRNA in the injured alveolus.


Asunto(s)
Lesión Pulmonar Aguda/terapia , Micropartículas Derivadas de Células/trasplante , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Lesión Pulmonar Aguda/microbiología , Lesión Pulmonar Aguda/patología , Animales , Endotoxinas/toxicidad , Escherichia coli/metabolismo , Humanos , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica
10.
Anesthesiology ; 121(5): 1099-121, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25211170

RESUMEN

Critically ill patients often suffer from multiple organ failures involving lung, kidney, liver, or brain. Genomic, proteomic, and metabolomic approaches highlight common injury mechanisms leading to acute organ failure. This underlines the need to focus on therapeutic strategies affecting multiple injury pathways. The use of adult stem cells such as mesenchymal stem or stromal cells (MSC) may represent a promising new therapeutic approach as increasing evidence shows that MSC can exert protective effects following injury through the release of promitotic, antiapoptotic, antiinflammatory, and immunomodulatory soluble factors. Furthermore, they can mitigate metabolomic and oxidative stress imbalance. In this work, the authors review the biological capabilities of MSC and the results of clinical trials using MSC as therapy in acute organ injuries. Although preliminary results are encouraging, more studies concerning safety and efficacy of MSC therapy are needed to determine their optimal clinical use. (ANESTHESIOLOGY 2014; 121:1099-121).


Asunto(s)
Enfermedad Crítica/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Traumatismo Múltiple/terapia , Humanos , Células Madre Mesenquimatosas/fisiología , Insuficiencia Multiorgánica/prevención & control , Insuficiencia Multiorgánica/terapia
11.
Anesthesiology ; 118(5): 1098-105, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23353794

RESUMEN

BACKGROUND: Postoperative cognitive decline can be reproduced in animal models. In a well-validated rat model of the Metabolic Syndrome, we sought to investigate whether surgery induced a more severe and persistent form of cognitive decline similar to that noted in preliminary clinical studies. METHODS: In rats that had been selectively bred for low and high exercise endurance, the low capacity runners (LCR) exhibited features of Metabolic Syndrome (obesity, dyslipidemia, insulin resistance, and hypertension). Tibial fracture surgery was performed under isoflurane anesthesia in LCR and high capacity runner (HCR) rats and cognitive function was assessed postoperatively in a trace-fear conditioning paradigm and Morris Water Maze; non-operated rats were exposed to anesthesia and analgesia (sham). Group sizes were n = 6. RESULTS: On postoperative D7, LCR rats had shorter freezing times than postoperative HCR rats. Five months postoperatively, LCR rats had a flatter learning trajectory and took longer to locate the submerged platform than postoperative HCR rats; dwell-time in the target quadrant in a probe trial was shorter in the postoperative LCR compared to HCR rats. LCR and HCR sham rats did not differ in any test. CONCLUSION: Postoperatively, LCR rats diverged from HCR rats exhibiting a greater decline in memory, acutely, with persistent learning and memory decline, remotely; this could not be attributed to changes in locomotor or swimming performance. This Metabolic Syndrome animal model of surgery-induced cognitive decline corroborates, with high fidelity, preliminary findings of postoperative cognitive dysfunction in Metabolic Syndrome patients.


Asunto(s)
Trastornos del Conocimiento/psicología , Síndrome Metabólico/psicología , Complicaciones Posoperatorias/psicología , Análisis de Varianza , Anestesia General , Animales , Trastornos del Conocimiento/complicaciones , Condicionamiento Psicológico , Señales (Psicología) , Miedo/psicología , Inmunidad/fisiología , Inflamación/patología , Masculino , Aprendizaje por Laberinto , Memoria a Largo Plazo/fisiología , Síndrome Metabólico/complicaciones , Actividad Motora/fisiología , Complicaciones Posoperatorias/etiología , Ratas , Carrera/fisiología , Percepción Espacial/fisiología , Natación/psicología , Fracturas de la Tibia/psicología , Fracturas de la Tibia/cirugía
12.
Respirology ; 18(5): 744-56, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23578018

RESUMEN

Acute lung injury (ALI) or acute respiratory distress syndrome remains a major cause of morbidity and mortality in hospitalized patients. The pathophysiology of ALI involves complex interactions between the inciting event, such as pneumonia, sepsis or aspiration, and the host immune response resulting in lung protein permeability, impaired resolution of pulmonary oedema, an intense inflammatory response in the injured alveolus and hypoxemia. In multiple preclinical studies, adult stem cells have been shown to be therapeutic due to both the ability to mitigate injury and inflammation through paracrine mechanisms and perhaps to regenerate tissue by virtue of their multi-potency. These characteristics have stimulated intensive research efforts to explore the possibility of using stem or progenitor cells for the treatment of lung injury. A variety of stem or progenitor cells have been isolated, characterized and tested experimentally in preclinical animal models of ALI. However, questions remain concerning the optimal dose, route and the adult stem or progenitor cell to use. Here, the current mechanisms underlying the therapeutic effect of stem cells in ALI as well as the questions that will arise as clinical trials for ALI are planned are reviewed.


Asunto(s)
Lesión Pulmonar Aguda/terapia , Células Madre Adultas/trasplante , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , Modelos Animales de Enfermedad , Humanos , Investigación con Células Madre
13.
Crit Care ; 16(3): R109, 2012 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-22731894

RESUMEN

INTRODUCTION: To evaluate the efficacy of probiotics in preventing nosocomial pneumonia in critically ill patients. METHODS: We searched PubMed, EMBASE, and the Web of Science for relevant studies. Two reviewers extracted data and reviewed the quality of the studies independently. The primary outcome was the incidence of nosocomial pneumonia. Study-level data were pooled using a random-effects model when I(2) was > 50% or a fixed-effects model when I(2) was < 50%. RESULTS: Twelve randomized controlled studies with a total of 1,546 patients were considered. Pooled analysis showed a statistically significant reduction in nosocomial pneumonia rates due to probiotics (odd ratio [OR]= 0.75, 95% CI 0.57 to 0.97, P = 0.03, I(2) = 46%). However, no statistically significant difference was found between groups regarding in-hospital mortality (OR = 0.93, 95% CI 0.50 to 1.74, P = 0.82, I(2) = 51%), intensive care unit mortality (OR = 0.84, 95% CI 0.55 to 1.29, P = 0.43, I(2) = 0%), duration of stay in the hospital (mean difference [MD] in days = -0.13, 95% CI -0.93 to 0.67, P = 0.75, I(2) = 46%), or duration of stay in the intensive care units (MD = -0.72, 95% CI -1.73 to 0.29, P = 0.16, I(2) = 68%). CONCLUSIONS: The use of probiotics was associated with a statistically significant reduction in the incidence of nosocomial pneumonia in critically ill patients. However, large, well-designed, randomized, multi-center trials are needed to confirm any effects of probiotics clinical endpoints such as mortality and length of ICU and hospital stay.


Asunto(s)
Enfermedad Crítica/terapia , Infección Hospitalaria/dietoterapia , Unidades de Cuidados Intensivos , Neumonía Asociada al Ventilador/dietoterapia , Probióticos/uso terapéutico , Enfermedad Crítica/epidemiología , Infección Hospitalaria/epidemiología , Humanos , Neumonía Asociada al Ventilador/epidemiología , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos
14.
Stem Cell Res Ther ; 13(1): 220, 2022 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-35619189

RESUMEN

BACKGROUND: Existing clinical studies supported the potential efficacy of mesenchymal stromal cells as well as derived exosomes in the treatment of COVID-19. We aimed to explore the safety and efficiency of aerosol inhalation of the exosomes derived from human adipose-derived MSCs (haMSC-Exos) in patients with COVID-19. METHODS: The MEXCOVID trial is a phase 2a single-arm, open-labelled, interventional trial and patients were enrolled in Jinyintan Hospital, Wuhan, China. Eligible 7 patients were assigned to receive the daily dose of haMSCs-Exos (2.0 × 108 nano vesicles) for consecutively 5 days. The primary outcomes included the incidence of prespecified inhalation-associated events and serious adverse events. We also observed the demographic data, clinical characteristics, laboratory results including lymphocyte count, levels of D-dimer and IL-6 as well as chest imaging. RESULTS: Seven severe COVID-19 related pneumonia patients (4 males and 3 females) were enrolled and received nebulized haMSC-Exos. The median age was 57 year (interquartile range (IQR), 43 year to 70 year). The median time from onset of symptoms to hospital admission and administration of nebulized haMSC-Exos was 30 days (IQR, 15 days to 40 days) and 54 d (IQR, 34 d to 69 d), respectively. All COVID-19 patients tolerated the haMSC-Exos nebulization well, with no evidence of prespecified adverse events or clinical instability during the nebulization or during the immediate post-nebulization period. All patients presented a slight increase of serum lymphocyte counts (median as 1.61 × 109/L vs. 1.78 × 109/L). Different degrees of resolution of pulmonary lesions after aerosol inhalation of haMSC-Exos were observed among all patients, more obviously in 4 of 7 patients. CONCLUSIONS: Our trial shows that a consecutive 5 days inhalation dose of clinical grade haMSC-Exos up to a total amount of 2.0 × 109 nano vesicles was feasible and well tolerated in seven COVID-19 patients, with no evidence of prespecified adverse events, immediate clinical instability, or dose-relevant toxicity at any of the doses tested. This safety profile is seemingly followed by CT imaging improvement within 7 days. Further trials will have to confirm the long-term safety or efficacy in larger population. TRIAL REGISTRATION: MEXCOVID, NCT04276987.


Asunto(s)
COVID-19 , Exosomas , Células Madre Mesenquimatosas , Tejido Adiposo , COVID-19/terapia , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto
15.
Respir Res ; 12: 158, 2011 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-22176972

RESUMEN

BACKGROUND: Only 10-15% of smokers develop chronic obstructive pulmonary disease (COPD) which indicates genetic susceptibility to the disease. Recent studies suggested an association between COPD and polymorphisms in CHRNA coding subunits of nicotinic acetylcholine receptor. Herein, we performed a meta-analysis to clarify the impact of CHRNA variants on COPD. METHODS: We searched Web of Knowledge and Medline from 1990 through June 2011 for COPD gene studies reporting variants on CHRNA. Pooled odds ratios (ORs) were calculated using the major allele or genotype as reference group. RESULTS: Among seven reported variants in CHRNA, rs1051730 was finally analyzed with sufficient studies. Totally 3460 COPD and 11437 controls from 7 individual studies were pooled-analyzed. A-allele of rs1051730 was associated with an increased risk of COPD regardless of smoking exposure (pooled OR = 1.26, 95% CI 1.18-1.34, p < 10⁻5). At the genotypic level, the ORs gradually increased per A-allele (OR = 1.27 and 1.50 for GA and AA respectively, p < 10⁻5). Besides, AA genotype exhibited an association with reduced FEV1% predicted (mean difference 3.51%, 95%CI 0.87-6.16%, p = 0.009) and increased risk of emphysema (OR 1.93, 95%CI 1.29-2.90, p = 0.001). CONCLUSIONS: Our findings suggest that rs1051730 in CHRNA is a susceptibility variant for COPD, in terms of both airway obstruction and parenchyma destruction.


Asunto(s)
Polimorfismo de Nucleótido Simple , Enfermedad Pulmonar Obstructiva Crónica/genética , Receptores Nicotínicos/genética , Estudios de Casos y Controles , Volumen Espiratorio Forzado , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Humanos , Pulmón/fisiopatología , Oportunidad Relativa , Fenotipo , Enfermedad Pulmonar Obstructiva Crónica/epidemiología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Enfisema Pulmonar/genética , Medición de Riesgo , Factores de Riesgo , Fumar/efectos adversos , Fumar/epidemiología
16.
BMC Infect Dis ; 11: 280, 2011 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-22014385

RESUMEN

BACKGROUND: Pleural infection is a common clinical problem. Its successful treatment depends on rapid diagnosis and early initiation of antibiotics. The measurement of soluble triggering receptor expressed in myeloid cells-1 (sTREM-1) level in pleural effusions has proven to be a valuable diagnostic tool for differentiating bacterial effusions from effusions of other etiologies. Herein, we performed a meta-analysis to assess the accuracy of pleural fluid sTREM-1 in the diagnosis of bacterial infection. METHODS: We searched Web of Knowledge and Medline from 1990 through March 2011 for studies reporting diagnostic accuracy data regarding the use of sTREM-1 in the diagnosis of bacterial pleural effusions. Pooled sensitivity and specificity and summary measures of accuracy and Q* were calculated. RESULTS: Overall, the sensitivity of sTREM-1was 78% (95% CI: 72%-83%); the specificity was 84% (95% CI: 80%-87%); the positive likelihood ratio was 6.0 (95% CI: 3.3-10.7); and the negative likelihood ratio was 0.22 (95% CI: 0.12-0.40). The area under the SROC curve for sTREM-1 was 0.92. Statistical heterogeneity and inconsistency were found for sensitivity (p = 0.015, χ2 = 15.73, I2 = 61.9%), specificity (p = 0.000, χ2 = 29.90, I2 = 79.9%), positive likelihood ratio (p = 0.000, χ2 = 33.09, I2 = 81.9%), negative likelihood ratio (p = 0.008, χ2 = 17.25, I2 = 65.2%), and diagnostic odds ratio (p = 0.000, χ2 = 28.49, I2 = 78.9%). A meta-regression analysis performed showed that the Quality Assessment of Diagnostic Accuracy Studies score (p = 0.3245; RDOR, 4.34; 95% CI, 0.11 to 164.01), the Standards for Reporting of Diagnostic Accuracy score (p = 0.3331; RDOR, 1.70; 95% CI, 0.44 to 6.52), lack of blinding (p = 0.7439; RDOR, 0.60; 95% CI, 0.01 to 33.80), and whether the studies were prospective or retrospective studies (p = 0.2068; RDOR, 7.44; 95% CI, 0.18 to 301.17) did not affect the test accuracy. A funnel plot for publication bias suggested a remarkable trend of publication bias. CONCLUSIONS: Our findings suggest that sTREM-1 has a good diagnostic accuracy and may provide a useful adjunctive tool for the diagnosis of bacterial pleural effusions. However, further studies are needed in order to identify any differences in the diagnostic performance of sTREM-1 of parapneumonic effusions and empyemas.


Asunto(s)
Infecciones Bacterianas/diagnóstico , Biomarcadores/análisis , Glicoproteínas de Membrana/análisis , Derrame Pleural , Receptores Inmunológicos/análisis , Humanos , Sensibilidad y Especificidad , Receptor Activador Expresado en Células Mieloides 1
17.
Mediators Inflamm ; 2011: 560194, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21785528

RESUMEN

Acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS), continue to be a major cause of morbidity and mortality in critically ill patients. The present therapeutic strategies for ALI/ARDS including supportive care, pharmacological treatments, and ventilator support are still controversial. More scientists are focusing on therapies involving stem cells, which have self-renewing capabilities and differentiate into multiple cell lineages, and, genomics therapy which has the potential to upregulate expression of anti-inflammatory mediators. Recently, the combination of cell and gene therapy which has been demonstrated to provide additive benefit has opened up a new chapter in therapeutic strategy and provides a basis for the development of an innovative approach for the prevention and treatment of ALI/ARDS.


Asunto(s)
Lesión Pulmonar Aguda/terapia , Terapia Genética/métodos , Síndrome de Dificultad Respiratoria/terapia , Trasplante de Células Madre , Lesión Pulmonar Aguda/mortalidad , Enfermedad Crítica/mortalidad , Humanos , Síndrome de Dificultad Respiratoria/mortalidad
18.
Clin Transl Med ; 11(1): e287, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33463070

RESUMEN

RATIONALE: The effects of mesenchymal stromal cells (MSCs) and MSC-derived extracellular vesicles (MSC EVs) on multidrug-resistant pseudomonas aeruginosa (MDR-PA)-induced pneumonia remain unclear. MATERIALS AND METHODS: MicroRNA array and RT-PCR were used to select the major microRNA in MSC EVs. Human peripheral blood monocytes were obtained and isolated from qualified patients. The crosstalk between MSCs/MSC EVs and macrophages in vitro was studied. MDR-PA pneumonia models were further established in C57BL/6 mice and MSC EVs or miR-466 overexpressing MSC EVs were intratracheally instilled. RESULTS: MiR-466 was highly expressed in MSC EVs. MSCs and miR-466 promoted macrophage polarization toward Type 2 phenotype through TIRAP-MyD88-NFκB axis. Moreover, cocultured macrophages with miR-466 overexpressing MSCs significantly increased the phagocytosis of macrophages. MSC EVs significantly reduced mortality and decreased influx of BALF neutrophils, proinflammatory factor levels, protein, and bacterial load in murine MDR-PA pneumonia. Administration of miR-466 overexpressing MSC EVs further alleviated the inflammatory severity. CONCLUSIONS: MSC-derived EVs containing high levels of miR-466 may partly participate in host immune responses to MDR-PA. Both MSCs and MSC EVs have therapeutic effects in treating MDR-PA-induced pneumonia.


Asunto(s)
Vesículas Extracelulares/metabolismo , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Neumonía/metabolismo , Pseudomonas aeruginosa , Animales , Modelos Animales de Enfermedad , Resistencia a Múltiples Medicamentos/genética , Vesículas Extracelulares/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neumonía/genética
19.
Microorganisms ; 9(6)2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-34072189

RESUMEN

Clinical evidence suggests that nebulized colistimethate sodium (CMS) has benefits for treating lower respiratory tract infections caused by multidrug-resistant Gram-negative bacteria (GNB). Colistin is positively charged, while CMS is negatively charged, and both have a high molecular mass and are hydrophilic. These physico-chemical characteristics impair crossing of the alveolo-capillary membrane but enable the disruption of the bacterial wall of GNB and the aggregation of the circulating lipopolysaccharide. Intravenous CMS is rapidly cleared by glomerular filtration and tubular excretion, and 20-25% is spontaneously hydrolyzed to colistin. Urine colistin is substantially reabsorbed by tubular cells and eliminated by biliary excretion. Colistin is a concentration-dependent antibiotic with post-antibiotic and inoculum effects. As CMS conversion to colistin is slower than its renal clearance, intravenous administration can lead to low plasma and lung colistin concentrations that risk treatment failure. Following nebulization of high doses, colistin (200,000 international units/24h) lung tissue concentrations are > five times minimum inhibitory concentration (MIC) of GNB in regions with multiple foci of bronchopneumonia and in the range of MIC breakpoints in regions with confluent pneumonia. Future research should include: (1) experimental studies using lung microdialysis to assess the PK/PD in the interstitial fluid of the lung following nebulization of high doses of colistin; (2) superiority multicenter randomized controlled trials comparing nebulized and intravenous CMS in patients with pandrug-resistant GNB ventilator-associated pneumonia and ventilator-associated tracheobronchitis; (3) non-inferiority multicenter randomized controlled trials comparing nebulized CMS to intravenous new cephalosporines/ß-lactamase inhibitors in patients with extensive drug-resistant GNB ventilator-associated pneumonia and ventilator-associated tracheobronchitis.

20.
Clin Transl Med ; 11(11): e563, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34841721

RESUMEN

BACKGROUND: Our previous study shows that Adipose tissue-derived mesenchymal stem cells (ASCs) are a promising strategy for cell-based therapy against pulmonary infection with Pseudomonas aeruginosa (P. aeruginosa), but the underlying mechanisms remain unclear. METHODS: cDNA microarray assay was performed to explore the transcriptome of ASCs primed by P. aeruginosa. Small interfering RNA (siRNA) was constructed to select the receptor candidates for P. aeruginosa recognition and granulocyte-macrophage colony-stimulating factor (GM-CSF) production in ASCs. The soluble protein chimeras containing the extracellular domain of human CD69 fused to the Fc region of human immunoglobulin IgG1 were used as a probe to validate the recognition of P. aeruginosa. The association between CD69 and extracellular regulated protein kinases 1/2 (ERK1/2) was explored via co-immunoprecipitation, siRNA, and inhibitor. The murine models of P. aeruginosa pneumonia treated with WT-ASCs, GM-CSF-/- -ASCs Cd69-/- -ASCs or Erk1-/- -ASCs were used to determine the role of GM-CSF, CD69, and ERK1 in ASCs against P. aeruginosa infection. RESULTS: We showed that C-type lectin receptor CD69 mediated the protective effects of ASCs partly through GM-CSF. CD69 could specifically recognize P. aeruginosa and regulate GM-CSF secretion of ASCs. CD69 regulated the production of GM-CSF via ERK1 in ASCs after P. aeruginosa infection. Moreover, the Administration of ASCs with deficiency of CD69 or ERK1 completely blocked its protective effects in a murine model of P. aeruginosa pneumonia. CONCLUSIONS: CD69 recognizes P. aeruginosa and further facilitates ERK1 activation, which plays a crucial role in ASCs-based therapy against P. aeruginosa pneumonia. CD69 may be a novel target molecule to improve ASCs-based therapy against P. aeruginosa infection.


Asunto(s)
Antígenos CD/farmacología , Antígenos de Diferenciación de Linfocitos T/farmacología , Células Madre Mesenquimatosas/metabolismo , Neumonía/terapia , Pseudomonas aeruginosa/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Lectinas Tipo C , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Neumonía/tratamiento farmacológico , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA