Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Stem Cells ; 41(5): 468-481, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36827175

ABSTRACT

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.


Subject(s)
Interferon Type I , Pneumonia , Mice , Animals , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Interleukin-7 , Membrane Proteins/genetics , Interferon Type I/metabolism , DNA/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Pneumonia/therapy
4.
Small ; 18(38): e2203035, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35988138

ABSTRACT

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.

5.
Stem Cell Res Ther ; 13(1): 220, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35619189

ABSTRACT

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.


Subject(s)
COVID-19 , Exosomes , Mesenchymal Stem Cells , Adipose Tissue , COVID-19/therapy , Female , Humans , Male , Middle Aged , Pilot Projects
6.
Clin Transl Med ; 11(11): e563, 2021 11.
Article in English | MEDLINE | ID: mdl-34841721

ABSTRACT

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.


Subject(s)
Antigens, CD/pharmacology , Antigens, Differentiation, T-Lymphocyte/pharmacology , Mesenchymal Stem Cells/metabolism , Pneumonia/therapy , Pseudomonas aeruginosa/drug effects , Animals , Disease Models, Animal , Lectins, C-Type , Mesenchymal Stem Cells/drug effects , Mice , Pneumonia/drug therapy , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/pathogenicity
7.
J Extracell Vesicles ; 10(10): e12134, 2021 08.
Article in English | MEDLINE | ID: mdl-34429860

ABSTRACT

Mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) turn out to be a promising source of cell-free therapy. Here, we investigated the biodistribution and effect of nebulized human adipose-derived MSC-EVs (haMSC-EVs) in the preclinical lung injury model and explored the safety of nebulized haMSC-EVs in healthy volunteers. DiR-labelled haMSC-EVs were used to explore the distribution of nebulized haMSC-EVs in the murine model. Pseudomonas aeruginosa-induced murine lung injury model was established, and survival rate, as well as WBC counts, histology, IL-6, TNF-α and IL-10 levels in bronchoalveolar lavage fluid (BALF) were measured to explore the optimal therapeutic dose of haMSC-EVs through the nebulized route. Twenty-four healthy volunteers were involved and received the haMSC-EVs once, ranging from 2 × 108 particles to 16 × 108 particles (MEXVT study, NCT04313647). Nebulizing haMSC-EVs improved survival rate to 80% at 96 h in P. aeruginosa-induced murine lung injury model by decreasing lung inflammation and histological severity. All volunteers tolerated the haMSC-EVs nebulization well, and no serious adverse events were observed from starting nebulization to the 7th day after nebulization. These findings suggest that nebulized haMSC-EVs could be a promising therapeutic strategy, offering preliminary evidence to promote the future clinical applications of nebulized haMSC-EVs in lung injury diseases.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Cytokines/metabolism , Drug Evaluation, Preclinical , Extracellular Vesicles/physiology , Lung Injury/therapy , Mesenchymal Stem Cells/physiology , Adolescent , Adult , Animals , Bronchoalveolar Lavage Fluid/chemistry , Disease Models, Animal , Female , Humans , Lung Injury/microbiology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Middle Aged , Patient Safety , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa , Survival Rate , Therapeutics/methods , Young Adult
8.
Microorganisms ; 9(6)2021 05 27.
Article in English | MEDLINE | ID: mdl-34072189

ABSTRACT

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.

9.
Curr Opin Infect Dis ; 34(2): 156-168, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33605620

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Pneumonia, Ventilator-Associated/drug therapy , Administration, Inhalation , Anti-Bacterial Agents/chemistry , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/physiology , Humans , Nebulizers and Vaporizers , Pneumonia, Ventilator-Associated/microbiology , Randomized Controlled Trials as Topic
10.
Clin Transl Med ; 11(1): e287, 2021 01.
Article in English | MEDLINE | ID: mdl-33463070

ABSTRACT

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.


Subject(s)
Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Pneumonia/metabolism , Pseudomonas aeruginosa , Animals , Disease Models, Animal , Drug Resistance, Multiple/genetics , Extracellular Vesicles/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Pneumonia/genetics
11.
Front Cell Infect Microbiol ; 10: 581535, 2020.
Article in English | MEDLINE | ID: mdl-33489931

ABSTRACT

Background: Pseudomonas aeruginosa (PA) is one of the most common Gram-negative bacteria causing hospital-acquired pulmonary infection, with high drug resistance and mortality. Therefore, it is urgent to introduce new non-antibiotic treatment strategies. Mesenchymal stem cells (MSCs), as important members of the stem cell family, were demonstrated to alleviate pathological damage in acute lung injury. However, the potential mechanism how MSC alleviate acute lung infection caused by PA remains unclear. Objective: The purpose of this study was to investigate the effects of Adipose-derived mesenchymal stem cells (ASCs) on acute pulmonary infections and the possible mechanisms how ASCs reduce pulmonary inflammation induced by PA. Methods: The therapeutic and mechanistic effects of ASCs on PA pulmonary infection were evaluated respectively in a murine model as well as in an in vitro model stimulated by PA and co-cultured with ASCs. Results: 1. ASCs treatment significantly reduced the bacterial load, inflammation of lung tissue and histopathological damage by PA. 2. PA infection mainly activated Nod-like receptor containing a caspase activating and recruitment domain 4 (NLRC4) inflammasome in the lung of mice. ASCs attenuated acute lung infection in mice by inhibiting NLRC4 inflammasome activation. 3. NLRC4-/- mice showed a significant improvement in survival rate and lung bacterial load after PA infection. 4. ASCs mainly increased expression and secretion of STC-1 in response to PA-stimulated NLRC4 inflammasome activation. Conclusions: PA infection attenuated macrophage phagocytosis through activation of NLRC4 inflammasome in macrophages, which eventually led to pulmonary inflammatory damage in mouse; ASCs reduced the activation of NLRC4 inflammasome in macrophages induced by PA infection, thereby increasing the phagocytic ability of macrophages, and ultimately improving lung tissue damage in mouse; ASCs may inhibit NLRC4 inflammasome through the secretion of STC-1.


Subject(s)
Inflammasomes , Mesenchymal Stem Cells , Animals , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins , Inflammasomes/metabolism , Lung/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Pseudomonas aeruginosa/metabolism
12.
Front Microbiol ; 10: 2198, 2019.
Article in English | MEDLINE | ID: mdl-31636610

ABSTRACT

RATIONALE: Although frequently retrieved in tracheal secretions of critically ill patients on mechanical ventilation, the existence of pneumonia caused by coagulase-negative staphylococci (CoNS) remains controversial. OBJECTIVE: To assess whether Staphylococcus haemolyticus (S. haemolyticus) inoculated in mice's trachea can infect normal lung parenchyma, increasing concentrations of S. haemolyticus were intratracheally administered in 221 immunocompetent mice. METHODS: Each animal received intratracheally phosphate-buffered saline (PBS) (n = 43) or live (n = 141) or inactivated (n = 37) S. haemolyticus at increasing load: 1.0 × 106, 1.0 × 107, and 1.0 × 108 colony forming units (CFU). Forty-three animals were sacrificed at 12 h and 178 were sacrificed at 36 h; 64 served for post-mortem lung histology, 157 served for pre-mortem bronchoalveolar lavage (BAL) analysis, and 42 served for post-mortem quantitative bacteriology of lung tissue. The distribution of biofilm-associated genes was investigated in the S. haemolyticus strain used in our in vivo experiment as well as among 19 other clinical S. haemolyticus strains collected from hospitals or nursing houses. MEASUREMENTS AND MAIN RESULTS: Intratracheal inoculation of 1.0 × 108 CFU live S. haemolyticus caused macroscopic and histological confluent pneumonia with significant increase in BAL white cell count, tumor necrosis factor-α (TNF-α), and macrophage inflammatory protein (MIP)-2. At 12 h, high concentrations of S. haemolyticus were identified in BAL. At 36 h, lung injury and BAL inflammation were less severe than at 12 h and moderate concentrations of species belonging to the oropharyngeal flora were identified in lung tissue. The inoculation of 1.0 × 106 and 1.0 × 107 CFU live S. haemolyticus caused histologic interstitial pneumonia and moderate BAL inflammation. Similar results were observed after inoculation of inactivated S. haemolyticus. Moreover, biofilm formation was a common phenotype in S. haemolyticus isolates. The low prevalence of the ica operon in our clinical S. haemolyticus strain collection indicated icaA and icaD independent-biofilm formation. CONCLUSION: In immunocompetent spontaneously breathing mice, inoculation of S. haemolyticus causes concentration-dependent lung infection that spontaneously recovers over time. icaA and icaD independent biofilm formation is a common phenotype in S. haemolyticus isolates.

13.
Clin Infect Dis ; 67(suppl_2): S146-S152, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30423049

ABSTRACT

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.


Subject(s)
Bacteria/isolation & purification , Candida/isolation & purification , Pneumonia, Ventilator-Associated/microbiology , Pneumonia, Ventilator-Associated/mortality , Aged , Aged, 80 and over , Anti-Bacterial Agents/therapeutic use , Bacteria/drug effects , Candida/drug effects , China/epidemiology , Drug Resistance, Multiple, Bacterial , Female , Hospitals, Teaching , Humans , Hypertension/complications , Length of Stay , Male , Middle Aged , Pneumonia, Ventilator-Associated/diagnosis , Prognosis , Retrospective Studies , Risk Factors
14.
ACS Appl Mater Interfaces ; 10(35): 29448-29456, 2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30088907

ABSTRACT

Metal-free bifunctional oxygen electrocatalysts are extremely critical to the advanced energy conversion devices, such as high energy metal-air batteries. Effective tuning of edge defects and electronic density on carbon materials via simple methods is especially attractive. In this work, a facile alkali activation method has been proposed to prepare carbon with large specific surface area and optimized porosity. In addition, subsequent nitrogen-doping leads to high pyridinic-N and graphitic-N contents and abundant edge defects, further enhancing electrochemical activities. Theoretical modeling via first-principles calculations has been conducted to correlate the electrocatalytic activities with their fundamental chemical structure of N doping and edge defect engineering. The metal-free product (NKCNPs-900) shows a high half-wave potential of 0.79 V (ORR). Furthermore, the assembled Zn-air batteries display excellent performance among carbon-based metal-free oxygen electrocatalysts, such as large peak power density up to 131.4 mW cm-2, energy density as high as 889.0 W h kg-1 at 4.5 mA cm-2, and remarkable discharge-charge cycles up to 575 times. Preliminarily, the rechargeable nonaqueous Li-air batteries were also investigated. Therefore, our work provides a low-cost, metal-free, and high-performance bifunctional carbon-based electrocatalyst for metal-air batteries.

15.
J Transl Int Med ; 6(1): 26-31, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29607301

ABSTRACT

The prevalence and microbial pattern reported for Community-acquired pneumonia (CAP) differ considerably and contemporary situation remains changing over time. We therefore searched both international and domestic databases for relevant references and pooled incidence of CAP and etiological distribution were estimated separately between children and adults groups. The results showed that CAP remained a major public health issue in China, with a relatively higher incidence than that reported in Western countries. Although pathogens were not detected in nearly half of patients, Mycoplasma pneumoniae remained to be the most frequently detected agent across age groups, the detection yield of which was lower than that reported from other countries. Notably, the incidence of influenza virus A in adults was almost four times higher than that in children while the prevalence of respiratory syncytial virus was much less common in adults than that in children. Despite some limitations, the value of this review, approaching to systematically review grey published data, is to sketch out the contemporary epidemiological and etiological situation of CAP in our country, which could be useful to help policymakers and clinicians make informed choices and to inspire future studies and surveillance.

16.
Int J Cancer ; 142(4): 769-778, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29023689

ABSTRACT

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.


Subject(s)
Lung Neoplasms/microbiology , Microbiota , Adult , Aged , Bronchoscopy , Case-Control Studies , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neisseria/isolation & purification , Neisseriaceae Infections/microbiology , Streptococcal Infections/microbiology , Streptococcus/isolation & purification
17.
Stem Cells ; 35(7): 1849-1859, 2017 07.
Article in English | MEDLINE | ID: mdl-28376568

ABSTRACT

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.


Subject(s)
Acute Lung Injury/prevention & control , Angiopoietin-1/genetics , Cell-Derived Microparticles/metabolism , Lung/metabolism , Mesenchymal Stem Cells/metabolism , RNA, Messenger/genetics , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Angiopoietin-1/antagonists & inhibitors , Angiopoietin-1/metabolism , Animals , Bronchoalveolar Lavage Fluid/chemistry , Capillary Permeability/genetics , Cell-Derived Microparticles/chemistry , Cell-Derived Microparticles/transplantation , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Lipopolysaccharides , Lung/pathology , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/genetics , Neutrophils/metabolism , Neutrophils/pathology , Primary Cell Culture , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
J Nat Med ; 71(1): 76-85, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27491743

ABSTRACT

Myrislignan is a natural compound with little pharmacological study. In our investigation, we investigated the effect of myrislignan in the induction of apoptosis in A549 cells in vitro and in vivo. Myrislignan inhibited the proliferation of A549 cells in a dose- and time-dependent manner assayed by MTT. In addition, Hoechst flow cytometry showed that myrislignan significantly induced apoptosis and cell cycle arrest in A549 cells. The apoptosis and anti-cell proliferation was mediated by the activation of mitogen-activated protein kinase and the inhibition of epidermal growth factor receptor signal pathway, change of mitochondrial membrane potential, the releasing of c-Myc, the downregulation of the level of the anti-apoptotic protein Bcl-2, and the upregulation of the level of the pro-apoptotic protein Bax. In conclusion, those results reveal a potential mechanism for the anti-cancer effect of myrislignan on human lung cancer, while suggesting that myrislignan may be a promising compound for the treatment of lung cancer.


Subject(s)
Biological Products/therapeutic use , Lignans/therapeutic use , Lung Neoplasms/drug therapy , A549 Cells , Apoptosis , Biological Products/pharmacology , Cell Line, Tumor , Cell Proliferation , Humans , Immunohistochemistry , Lignans/pharmacology , Signal Transduction , Xenograft Model Antitumor Assays
19.
Expert Opin Biol Ther ; 16(7): 859-71, 2016 07.
Article in English | MEDLINE | ID: mdl-27011289

ABSTRACT

INTRODUCTION: Acute respiratory distress syndrome is a major cause of respiratory failure in critically ill patients. Despite extensive research into its pathophysiology, mortality remains high. No effective pharmacotherapy exists. Based largely on numerous preclinical studies, administration of mesenchymal stem or stromal cell (MSC) as a therapeutic for acute lung injury holds great promise, and clinical trials are currently underway. However, concern for the use of stem cells, specifically the risk of iatrogenic tumor formation, remains unresolved. Accumulating evidence now suggest that novel cell-free therapies including MSC-derived conditioned medium and extracellular vesicles released from MSCs might constitute compelling alternatives. AREAS COVERED: The current review summarizes the preclinical studies testing MSC conditioned medium and/or MSC extracellular vesicles as treatment for acute lung injury and other inflammatory lung diseases. EXPERT OPINION: While certain logistical obstacles limit the clinical applications of MSC conditioned medium such as the volume required for treatment, the therapeutic application of MSC extracellular vesicles remains promising, primarily due to ability of extracellular vesicles to maintain the functional phenotype of the parent cell. However, utilization of MSC extracellular vesicles will require large-scale production and standardization concerning identification, characterization and quantification.


Subject(s)
Acute Lung Injury/therapy , Extracellular Vesicles/physiology , Extracellular Vesicles/transplantation , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Animals , Culture Media, Conditioned , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Lung Diseases/immunology , Lung Diseases/metabolism , Lung Diseases/therapy
20.
Int J Antimicrob Agents ; 46(6): 603-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26607337

ABSTRACT

Colistin has been used to treat nosocomial pneumonia (NP) caused by multidrug-resistant (MDR) Gram-negative bacteria (GNB) via different administration routes. Whether patients may benefit from aerosolised colistin as adjunctive treatment was contradictory. We aimed to clarify the safety and efficacy of administering aerosolised and intravenous (IV-AS) colistin versus intravenous (IV) colistin alone in patients with NP caused by MDR-GNB. Two reviewers independently evaluated and extracted data from PubMed, EMBASE and Cochrane databases. Primary outcomes were clinical response rate, all-cause mortality (ICU or hospital), microbiological eradication and nephrotoxicity. Pooled odds ratios (ORs) were calculated and significance was determined by the Z test. Nine eligible studies involving 672 participants were included. The overall clinical response rate (improvement and cure) was significantly higher in the IV-AS group than that in the IV group [OR=1.81, 95% confidence interval (CI) 1.30-2.53; P=0.0005]. Patients treated with IV-AS colistin showed a higher rate of pathogen eradication (OR=1.66, 95% CI 1.11-2.49; P=0.01) and lower all-cause mortality compared with IV colistin (OR=0.69, 95% CI 0.50-0.95; P=0.02). Nephrotoxicity did not differ significantly between IV-AS and IV groups (five studies; 383 patients) (OR=1.11, 95% CI 0.69-1.80; P=0.67). These data indicate that IV-AS colistin has additional benefits compared with IV colistin alone. Clinicians should be encouraged to give combined administration routes in critically ill patients with NP caused by MDR-GNB.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Colistin/administration & dosage , Colistin/therapeutic use , Gram-Negative Bacterial Infections/drug therapy , Pneumonia, Ventilator-Associated/drug therapy , Administration, Inhalation , Administration, Intravenous , Colistin/adverse effects , Critical Illness , Drug Resistance, Multiple, Bacterial , Gram-Negative Bacteria/drug effects , Gram-Negative Bacterial Infections/microbiology , Humans , Nasal Sprays , Pneumonia, Ventilator-Associated/microbiology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...