Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
ERJ Open Res ; 10(3)2024 May.
Article En | MEDLINE | ID: mdl-38770009

Background: In China, the prevalence of severe asthma with eosinophilic phenotype is rising, yet treatment options are limited. Mepolizumab is the first targeted biologic therapy for eosinophilic-driven disease in China. This study (clinicaltrials.gov identifier NCT03562195) evaluated efficacy and safety of mepolizumab in Chinese patients with severe asthma. Methods: The phase III, multicentre, randomised, placebo-controlled, double-blind, parallel-group study enrolled patients aged ≥12 years with severe asthma, with two or more exacerbations in the previous year, and on inhaled corticosteroids plus at least one controller medication. Following a 1-4-week run-in, patients were randomised 1:1 to mepolizumab 100 mg or placebo subcutaneously every 4 weeks for 52 weeks. The primary end-point was annualised rate of clinically significant exacerbations (CSEs) through week 52. Secondary end-points were time to first CSE, frequency of CSEs requiring hospitalisation/emergency department visits or hospitalisation over 52 weeks, mean change in St George's Respiratory Questionnaire (SGRQ) total score and pre-bronchodilator forced expiratory volume in 1 s (FEV1) at week 52; safety was evaluated. Results: The modified intention-to-treat population included 300 patients. At week 52 with mepolizumab versus placebo, annualised rate of CSEs was 65% lower (0.45 versus 1.31 events per year; rate ratio 0.35, 95% CI 0.24-0.50; p<0.001); time to first CSE longer (hazard ratio 0.38, 95% CI 0.26-0.56; p<0.001) and number of CSEs requiring hospitalisation/emergency department visit lower (rate ratio 0.30, 95% CI 0.12-0.77; p=0.012). From baseline to week 52, SGRQ score improved (p=0.001) and pre-bronchodilator FEV1 increased (p=0.006). Incidence of adverse events was similar between treatment groups. Conclusion: Mepolizumab provided clinical benefits to patients with severe asthma in China and showed a favourable benefit-risk profile.

2.
Phys Rev Lett ; 132(11): 116201, 2024 Mar 15.
Article En | MEDLINE | ID: mdl-38563913

Intriguing topological polar structures in oxide nanofilms have drawn growing attention owing to their immense potential applications in nanoscale electronic devices. Here, we report a novel route to mechanically manipulate polar structures via flexoelectricity in wrinkled thin films. Our results present a flexoelectric polar transition from a nonpolar state to uniaxial polar stripes, biaxial meronlike or antimeronlike polar structures, and polar labyrinths by varying wrinkle morphologies. The evolution mechanisms and the outstanding mechanical tunability of these flexoelectric polar patterns were investigated theoretically and numerically. This strategy based on flexoelectricity for generating nontrivial polar structures will no longer rely on the superlattice structure and can be widely applicable to all centrosymmetric or noncentrosymmetric materials, providing a broader range of material and structure candidates for polar topologies.

3.
Nanomaterials (Basel) ; 14(8)2024 Apr 19.
Article En | MEDLINE | ID: mdl-38668209

Lenses have been a cornerstone of optical systems for centuries; however, they are inherently limited by the laws of physics, particularly in terms of size and weight. Because of their characteristic light weight, small size, and subwavelength modulation, metalenses have the potential to miniaturize and integrate imaging systems. However, metalenses still face the problem that chromatic aberration affects the clarity and accuracy of images. A high-quality image system based on the end-to-end joint optimization of a neural network and an achromatic metalens is demonstrated in this paper. In the multi-scale encoder-decoder network, both the phase characteristics of the metalens and the hyperparameters of the neural network are optimized to obtain high-resolution images. The average peak-signal-to-noise ratio (PSNR) and average structure similarity (SSIM) of the recovered images reach 28.53 and 0.83. This method enables full-color and high-performance imaging in the visible band. Our approach holds promise for a wide range of applications, including medical imaging, remote sensing, and consumer electronics.

4.
Am J Respir Crit Care Med ; 208(4): 435-441, 2023 08 15.
Article En | MEDLINE | ID: mdl-37315325

Rationale: The CAPTURE tool (Chronic Obstructive Pulmonary Disease [COPD] Assessment in Primary Care to Identify Undiagnosed Respiratory Disease and Exacerbation Risk) was developed to identify patients with undiagnosed COPD with an FEV1 <60% predicted or risk of exacerbation as treatment criteria. Objectives: To test the ability of CAPTURE to identify patients requiring treatment because of symptoms or risk of exacerbation or hospitalization. Methods: Data were from COMPASS (Clinical, Radiological and Biological Factors Associated with Disease Progression, Phenotypes and Endotypes of COPD in China), a prospective study of COPD, chronic bronchitis without airflow limitation (postbronchodilator FEV1/FVC ratio ≥0.70), and healthy never-smokers. CAPTURE was tested as questions alone and with peak expiratory flow measurement. Sensitivity, specificity, and positive and negative predicted values (PPV and NPV) were calculated for COPD Assessment Test (CAT) scores ⩾10 versus <10, modified Medical Research Council (mMRC) scores ⩾2 versus <2, and at least one moderate exacerbation or hospitalization in the previous year versus none. Measurements and Main Results: Patients with COPD (n = 1,696) had a mean age of 65 ± 7.5 years, and 90% were male, with a postbronchodilator FEV1 of 66.5 ± 20.1% predicted. Control participants (n = 307) had a mean age of 60.2 ± 7.0 years, and 65% were male, with an FEV1/FVC ratio of 0.78 ± 0.04. CAPTURE using peak expiratory flow showed the best combination of sensitivity and specificity. Sensitivity and specificity were 68.5% and 64.0%, respectively, to detect a CAT score ⩾10; 85.6% and 61.0% to detect an mMRC score ⩾2; 63.5% and 55.6% to detect at least one moderate exacerbation; and 70.2% and 59.4% to detect at least one hospitalization. PPVs ranged from 15.6% (moderate exacerbations) to 47.8% (CAT score). NPVs ranged from 80.8% (CAT score) to 95.6% (mMRC score). Conclusions: CAPTURE has good sensitivity to identify patients with COPD who may require treatment because of increased symptoms or risk of exacerbations or hospitalization, including those with an FEV1 >60% predicted. High NPV values show that CAPTURE can also exclude those who may not require treatment. Clinical trial registered with www.clinicaltrials.gov (NCT04853225).


Pulmonary Disease, Chronic Obstructive , Male , Female , Humans , Prospective Studies , Forced Expiratory Volume , Lung , Sensitivity and Specificity , Disease Progression
5.
Nanomaterials (Basel) ; 12(23)2022 Dec 03.
Article En | MEDLINE | ID: mdl-36500921

Metalenses composed of a large number of subwavelength nanostructures provide the possibility for the miniaturization and integration of the optical system. Broadband polarization-insensitive achromatic metalenses in the visible light spectrum have attracted researchers because of their wide applications in optical integrated imaging. This paper proposes a polarization-insensitive achromatic metalens operating over a continuous bandwidth from 470 nm to 700 nm. The silicon nitride nanopillars of 488 nm and 632.8 nm are interleaved by Fresnel zone spatial multiplexing method, and the particle swarm algorithm is used to optimize the phase compensation. The maximum time-bandwidth product in the phase library is 17.63. The designed focal length can be maintained in the visible light range from 470 nm to 700 nm. The average focusing efficiency reaches 31.71%. The metalens can achieve broadband achromatization using only one shape of nanopillar, which is simple in design and easy to fabricate. The proposed metalens is expected to play an important role in microscopic imaging, cameras, and other fields.

6.
Sensors (Basel) ; 22(21)2022 Oct 24.
Article En | MEDLINE | ID: mdl-36365833

In the field of ultra high accuracy inertial measurement unit (IMU), pendulous integrating gyroscopic accelerometer (PIGA) has become a research hot spot due to its high-end performance. However, PIGA is sensitive to angular velocity, and the calibration process of PIGA-based IMU will be very complicated, which makes online self-calibration difficult to implement. To solve the above problems, we proposed an online self-calibration method utilizing angular velocity observation. The main contributions of this study are twofold: (1) An error analysis of PIGA is conducted in this paper, and the error model has also been simplified to suit the self-calibration model. (2) An improved online self-calibration method utilizing angular observation based on a simplified PIGA error model is proposed in this study. Experimental results show that the self-calibration method proposed in this study can improve the PIGA online calibration accuracy effectively (with the accuracy within 0.02 m/s/pulse), which can improve the dynamic accuracy of the PIGA.

7.
Stem Cell Res Ther ; 13(1): 496, 2022 10 08.
Article En | MEDLINE | ID: mdl-36209115

BACKGROUND: Microvesicles (MVs) derived from human bone marrow mesenchymal stem cell (MSC) were demonstrated to restore lung protein permeability and attenuate acute lung injury. In our previous study, we found that MSC MV increased sphingosine-1-phosphate (S1P) kinase1 mRNA levels in injured human lung microvascular endothelial cells (HLMVEC) significantly. However, the role of S1P signaling in MSC MV to restore lung protein permeability is unknown. METHODS: In this study, we hypothesized that MSC MV might restore lung permeability in part through increasing intracellular S1P signaling pathway in injured HLMVEC independent of S1P receptors. We used the transwell co-culture system to study the effect of MSC MV on protein permeability of Lipopolysaccharide (LPS) damaged HLMVEC. RESULTS: Our results showed that LPS significantly increased the permeability of HLMVEC to FITC-dextran (70 kDa) within 24 h. MSC MV restores this permeability and, to a large extent, prevents the cytoskeleton protein F-actin from recombining into "actin stress fibers," and restores the positions of tight junctions and adhesion junctions in the damaged HLMVEC. This therapeutic effect of MSC MV was related to the increase in the S1P level in injured HLMVEC and was not eliminated when adding the antagonist of S1P receptor, suggesting that MSC MV to restore lung permeability was independent of S1P receptors on HLMVEC. Laser confocal further observed that Ca2+ mobilization and Rac1 activation in LPS injured HLMVEC were increased in parallel with the increase in intracellular S1P level after MSC MV treatment. CONCLUSIONS: In short, MSC MV partially restored protein permeability across HLMVEC through the intracellular S1P signaling pathway independent of S1P receptor-1.


Lipopolysaccharides , Mesenchymal Stem Cells , Sphingosine-1-Phosphate Receptors/metabolism , Actins/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Humans , Lipopolysaccharides/pharmacology , Lung/metabolism , Lysophospholipids , Mesenchymal Stem Cells/metabolism , Permeability , RNA, Messenger/metabolism , Signal Transduction , Sphingosine/analogs & derivatives
10.
Dermatol Ther ; 35(4): e15303, 2022 04.
Article En | MEDLINE | ID: mdl-34984792

Chronic spontaneous urticaria (CSU) is characterized by the spontaneous development of wheals, itching, and/or angioedema, for ≥6 weeks. In China, non-sedating H1-antihistamines (H1AH) are the recommended first-line treatment, with escalation up to 4× the standard dose in symptomatic patients to achieve control. Treatment options for Chinese patients who remain symptomatic on H1AH treatment are limited. This 20-week randomized, double blind, placebo-controlled, parallel-group study investigated the efficacy and safety of omalizumab as an add-on therapy for the treatment of patients with CSU who remained symptomatic despite H1AH treatment in China. Adult patients (N = 418) diagnosed with refractory CSU for ≥6 months were randomized (2:2:1) to receive omalizumab 300 mg (OMA300), omalizumab 150 mg (OMA150) or placebo, subcutaneously, every 4 weeks. Primary outcome was change from baseline to week 12 in weekly itch severity score (ISS7). Safety was assessed by rates of adverse events (AEs). Demographic and disease characteristics at baseline were comparable across treatment groups. At week 12, statistically significant greater decreases from baseline were observed in ISS7 with OMA300 (least square mean difference [LSM]: -4.23; 95% confidence interval [CI]: -5.70, -2.77; p < 0.001) and OMA150 (LSM: -3.79; 95% CI: -5.24, -2.33; p < 0.001) versus placebo. Incidence of treatment-emergent AEs over 20 weeks was slightly higher with OMA300 (71.3%) compared to OMA150 and placebo groups (64.7% and 63.9%, respectively). The incidences of serious AEs were balanced between groups. This study demonstrated the efficacy and safety of omalizumab in Chinese adult patients with CSU who remained symptomatic despite H1AH therapy.


Anti-Allergic Agents , Chronic Urticaria , Urticaria , Adult , Anti-Allergic Agents/adverse effects , Chronic Disease , Chronic Urticaria/diagnosis , Chronic Urticaria/drug therapy , Histamine H1 Antagonists , Humans , Omalizumab/adverse effects , Treatment Outcome , Urticaria/chemically induced , Urticaria/diagnosis , Urticaria/drug therapy
11.
Front Public Health ; 9: 720512, 2021.
Article En | MEDLINE | ID: mdl-34616706

Ensuring the well-being of persons with disabilities (PWDs) is a priority in the public sector during the coronavirus disease 2019 (COVID-19) pandemic. To contain this unprecedented public crisis in China, a set of nationwide anti-epidemic discourse systems centered on war metaphors has guided the epidemic's prevention and control. While the public is immersed in the joy brought by the stage victory, most ignore the situation of the disadvantaged PWDs. Accordingly, this study adopts and presents a qualitative research method to explore the impact of war metaphors on PWDs. The results showed that while there was some formal and informal support for PWDs during this period, they were increasingly marginalized. Owing to the lack of a disability lens and institutional exclusion, PWDs were placed on the margins of the epidemic prevention and control system like outsiders. Affected by pragmatism under war metaphors, PWDs are regarded as non-contributory or inefficient persons; therefore, they are not prioritized and are thus placed into a state of being voiceless and invisible. This research can provide inspiration for improving public services for PWDs in the context of COVID-19.


COVID-19 , Disabled Persons , China/epidemiology , Humans , Metaphor , Pandemics/prevention & control , SARS-CoV-2
12.
J Immunol ; 203(7): 1961-1972, 2019 10 01.
Article En | MEDLINE | ID: mdl-31451675

Human mesenchymal stem cell (MSC) extracellular vesicles (EV) can reduce the severity of bacterial pneumonia, but little is known about the mechanisms underlying their antimicrobial activity. In the current study, we found that bacterial clearance induced by MSC EV in Escherichia coli pneumonia in C57BL/6 mice was associated with high levels of leukotriene (LT) B4 in the injured alveolus. More importantly, the antimicrobial effect of MSC EV was abrogated by cotreatment with a LTB4 BLT1 antagonist. To determine the role of MSC EV on LT metabolism, we measured the effect of MSC EV on a known ATP-binding cassette transporter, multidrug resistance-associated protein 1 (MRP1), and found that MSC EV suppressed MRP1 mRNA, protein, and pump function in LPS-stimulated Raw264.7 cells in vitro. The synthesis of LTB4 and LTC4 from LTA4 are competitive, and MRP1 is the efflux pump for LTC4 Inhibition of MRP1 will increase LTB4 production. In addition, administration of a nonspecific MRP1 inhibitor (MK-571) reduced LTC4 and subsequently increased LTB4 levels in C57BL/6 mice with acute lung injury, increasing overall antimicrobial activity. We previously found that the biological effects of MSC EV were through the transfer of its content, such as mRNA, microRNA, and proteins, to target cells. In the current study, miR-145 knockdown abolished the effect of MSC EV on the inhibition of MRP1 in vitro and the antimicrobial effect in vivo. In summary, MSC EV suppressed MRP1 activity through transfer of miR-145, thereby resulting in enhanced LTB4 production and antimicrobial activity through LTB4/BLT1 signaling.


Acute Lung Injury , Escherichia coli Infections , Escherichia coli/immunology , Extracellular Vesicles , Mesenchymal Stem Cells/immunology , Pneumonia, Bacterial , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Acute Lung Injury/therapy , Animals , Escherichia coli Infections/immunology , Escherichia coli Infections/therapy , Extracellular Vesicles/immunology , Extracellular Vesicles/pathology , Extracellular Vesicles/transplantation , Humans , Leukotriene B4/immunology , Leukotriene C4/immunology , Lung/immunology , Lung/pathology , Male , Mesenchymal Stem Cells/pathology , Mice , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Multidrug Resistance-Associated Proteins/immunology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/therapy , Propionates/pharmacology , Quinolines/pharmacology , RAW 264.7 Cells
13.
Thorax ; 74(1): 43-50, 2019 01.
Article En | MEDLINE | ID: mdl-30076187

BACKGROUND: We previously reported that microvesicles (MVs) released by human mesenchymal stem cells (MSC) were as effective as the cells themselves in both Escherichia coli lipopolysaccharide and live bacteria-induced acute lung injury (ALI) mice models. However, it remained unclear whether the biological effect of MSC MV can be applied to human ALI. METHODS: In the current study, we tested the therapeutic effects of MSC MVs in a well-established ex vivo perfused human model of bacterial pneumonia. Using human donor lungs not used for transplantation, we instilled E. coli bacteria intrabronchially and, 1 hour later, administered MSC MVs into the perfusate as therapy. RESULTS: After 6 hours, instillation of E. coli bacteria caused influx of inflammatory cells, which resulted in significant inflammation, lung protein permeability and pulmonary oedema formation. Administration of MSC MV significantly increased alveolar fluid clearance and reduced protein permeability and numerically lowered the bacterial load in the injured alveolus. The beneficial effect on bacterial killing was more pronounced with pretreatment of MSCs with a Toll-like receptor 3 agonist, polyinosinic:polycytidylic acid (Poly (I:C)), prior to the isolation of MVs. Isolated human alveolar macrophages had increased antimicrobial activity with MSC MV treatment in vitro as well. Although oxygenation and lung compliance levels were similar between injury and treatment groups, administration of MSC MVs numerically decreased median pulmonary artery pressure at 6 hours. CONCLUSIONS: In summary, MSC MVs increased alveolar fluid clearance and reduced lung protein permeability, and pretreatment with Poly (I:C) enhanced the antimicrobial activity of MVs in an ex vivo perfused human lung with severe bacteria pneumonia.


Acute Lung Injury/physiopathology , Acute Lung Injury/therapy , Cell- and Tissue-Based Therapy , Cell-Derived Microparticles , Escherichia coli Infections/complications , Mesenchymal Stem Cells , Pneumonia, Bacterial/complications , Proteins/metabolism , Pulmonary Alveoli/metabolism , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Adult , Aged , Arterial Pressure , Bacterial Load , Cell-Derived Microparticles/drug effects , Female , Humans , Interferon Inducers/pharmacology , Leukocyte Count , Lung Compliance , Male , Mesenchymal Stem Cells/drug effects , Middle Aged , Neutrophils , Organ Culture Techniques , Oxygen/metabolism , Permeability , Poly I-C/pharmacology , Pulmonary Alveoli/microbiology , Pulmonary Alveoli/pathology , Pulmonary Artery , Pulmonary Edema/microbiology , Pulmonary Edema/therapy , Toll-Like Receptor 3/agonists , Tumor Necrosis Factor-alpha/metabolism
14.
Stem Cells Transl Med ; 7(10): 721-730, 2018 10.
Article En | MEDLINE | ID: mdl-30133167

Although mesenchymal stem cells (MSCs) transplantation has been shown to promote the lung respiration in acute lung injury (ALI) in vivo, its overall restorative capacity appears to be restricted mainly because of low retention in the injured lung. Angiotensin II (Ang II) are upregulated in the injured lung. Our previous study showed that Ang II increased MSCs migration via Ang II type 2 receptor (AT2R). To determine the effect of AT2R in MSCs on their cell migration after systemic injection in ALI mice, a human AT2R expressing lentiviral vector and a lentivirus vector carrying AT2R shRNA were constructed and introduced into human bone marrow MSCs. A mouse model of lipopolysaccharide-induced ALI was used to investigate the migration of AT2R-regulated MSCs and the therapeutic potential in vivo. Overexpression of AT2R dramatically increased Ang II-enhanced human bone marrow MSC migration in vitro. Moreover, MSC-AT2R accumulated in the damaged lung tissue at significantly higher levels than control MSCs 24 and 72 hours after systematic MSC transplantation in ALI mice. Furthermore, MSC-AT2R-injected ALI mice exhibited a significant reduction of pulmonary vascular permeability and improved the lung histopathology and had additional anti-inflammatory effects. In contrast, there were less lung retention in MSC-ShAT2R-injected ALI mice compared with MSC-Shcontrol after transplantation. Thus, MSC-ShAT2R-injected group exhibited a significant increase of pulmonary vascular permeability and resulted in a deteriorative lung inflammation. Our results demonstrate that overexpression of AT2R enhance the migration of MSCs in ALI mice and may provide a new therapeutic strategy for ALI. Stem Cells Translational Medicine 2018;7:721-730.


Acute Lung Injury/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Receptor, Angiotensin, Type 2/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cell Movement , Cytokines/analysis , Disease Models, Animal , Leukocyte Count , Lipopolysaccharides/toxicity , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Neutrophils/cytology , Receptor, Angiotensin, Type 2/genetics
15.
Stem Cells Transl Med ; 7(8): 615-624, 2018 08.
Article En | MEDLINE | ID: mdl-29737632

Our previous study demonstrated that mesenchymal stem cell (MSC) microvesicles (MV) reduced lung inflammation, protein permeability, and pulmonary edema in endotoxin-induced acute lung injury in mice. However, the underlying mechanisms for restoring lung protein permeability were not fully understood. In this current study, we hypothesized that MSC MV would restore protein permeability across injured human lung microvascular endothelial cells (HLMVEC) in part through the transfer of angiopoietin-1 (Ang1) mRNA to the injured endothelium. A transwell coculture system was used to study the effect of MSC MV on protein permeability across HLMVECs injured by cytomix, a mixture of IL-1ß, TNF-α, and IFN-γ (50 ng/ml). Our result showed that cytomix significantly increased permeability to FITC-dextran (70 kDa) across HLMVECs over 24 hours. Administration of MSC MVs restored this permeability in a dose dependent manner, which was associated with an increase in Ang1 mRNA and protein secretion in the injured endothelium. This beneficial effect was diminished when MSC MV was pretreated with an anti-CD44 antibody, suggesting that internalization of MV into the HLMVEC was required for the therapeutic effect. Fluorescent microscopy showed that MSC MV largely prevented the reorganization of cytoskeleton protein F-actin into "actin stress fiber" and restored the location of the tight junction protein ZO-1 and adherens junction protein VE-cadherin in injured HLMVECs. Ang1 siRNA pretreatment of MSC MV prior to administration to injured HLMVECs eliminated the therapeutic effect of MV. In summary, MSC MVs restored protein permeability across HLMVEC in part by increasing Ang1 secretion by injured HLMVEC. Stem Cells Translational Medicine 2018;7:615-624.


Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Actins/metabolism , Angiopoietin-1/antagonists & inhibitors , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Antibodies/immunology , Antibodies/pharmacology , Antigens, CD/metabolism , Cadherins/metabolism , Cells, Cultured , Coculture Techniques , Cytokines/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Hyaluronan Receptors/immunology , Lung/cytology , Mesenchymal Stem Cells/cytology , Microscopy, Fluorescence , Microvessels/cytology , Protein Transport/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Zonula Occludens-1 Protein/metabolism
16.
Sci Rep ; 8(1): 204, 2018 01 09.
Article En | MEDLINE | ID: mdl-29317710

Great interest has been shown in mesenchymal stem cell (MSC) therapy in a wide variety of clinical domains. However, the therapeutic efficiency depends on the proliferation and migration of MSCs. Chemokine receptors are involved in regulating the proliferation and migration to the specific organs of MSCs in different microenvironments. CXC receptor seven (CXCR7), a newly discovered Chemokine ligand 12 (CXCL12) receptor, has organ specificity for tumour migration. We hypothesized that CXCR7 expression affects proliferation and migration of MSCs. In present study, we constructed long-term and stable mMSCs lines overexpressing and suppressing CXCR7 modifications with lentiviral vectors. The transduction efficiencies, mRNA and protein expression of CXCR7 were significantly regulated. CXCR7 gene overexpression promoted mMSCs proliferation and migration, whereas suppressing CXCR7 had the opposite effect. Additional CXCL12 improved the vertical migration of mMSCs. The overexpression of CXCR7 increased the MSC-secreted CXCL12, VCAM-1, CD44 and MMP2 levels, which contributed to the improvement of mMSC proliferation and migration. Therefore, overexpressing CXCR7 improved the proliferation and migration of mMSCs, which may be attributable to the CXCL12 secreted by MSCs, leading to a positive feedback loop for CXCL12/CXCR7 axis. Our results may provide a potential method for improving the treatment effectiveness of mMSCs by overexpressing CXCR7.


Cell Movement , Cell Proliferation , Chemokine CXCL12/metabolism , Mesenchymal Stem Cells/metabolism , Receptors, CXCR/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Receptors, CXCR/genetics , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
17.
Stem Cell Res Ther ; 8(1): 211, 2017 09 29.
Article En | MEDLINE | ID: mdl-28969681

BACKGROUND: Mesenchymal stem cells microvesicles (MSC-MVs) stabilize endothelial barrier function in acute lung injury (ALI); however, the detailed mechanism remains to be further defined. Hepatocyte growth factor (HGF), which is derived from MSC-MVs, might have a key role in the restoration of endothelial barrier function by MSC-MVs. METHODS: MSCs with lentiviral vector-mediated HGF gene knockdown (siHGF-MSC) were generated. A co-culture model of pulmonary microvascular endothelial cells and MSC-MVs collected from MSCs or siHGF-MSCs after 24 h of hypoxic culture was utilized. Then, endothelial paracellular and transcellular permeabilities were detected. VE-cadherin, and occludin protein expression in the endothelial cells was measured using Western blot. Endothelial cell proliferation was analysed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) assay. Endothelial cell apoptosis was analysed using TUNEL assay. Finally, IL-6 and IL-10 production was determined via an enzyme-linked immunosorbent assay (ELISA). RESULTS: Treatment with MSC-MVs significantly decreased LPS-induced endothelial paracellular and transcellular permeabilities, and the effect was significantly inhibited after HGF gene knockdown in MSC-MVs. Furthermore, treatment with MSC-MVs increased the expression of the endothelial intercellular junction proteins VE-cadherin and occludin. Treatment with MSC-MVs also decreased endothelial apoptosis and induced endothelial cell proliferation. Finally, the treatment reduced IL-6 production and increased IL-10 production in the conditioned media of endothelial cells. However, the effects of the treatment with MSC-MVs were inhibited after HGF gene knockdown. CONCLUSIONS: MSC-MVs protect the barrier functions of pulmonary microvascular endothelial cells, which can be partly attributed to the presence of HGF in the MSC-MVs.


Capillary Permeability/genetics , Cell-Derived Microparticles/chemistry , Endothelial Cells/metabolism , Hepatocyte Growth Factor/genetics , Intercellular Junctions/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cadherins/genetics , Cadherins/metabolism , Capillary Permeability/drug effects , Cell Hypoxia , Cell Proliferation , Cell-Derived Microparticles/metabolism , Coculture Techniques , Culture Media, Conditioned/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Gene Expression , Hepatocyte Growth Factor/antagonists & inhibitors , Hepatocyte Growth Factor/metabolism , Intercellular Junctions/drug effects , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Occludin/genetics , Occludin/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
18.
Stem Cell Res Ther ; 8(1): 164, 2017 07 12.
Article En | MEDLINE | ID: mdl-28697804

BACKGROUND: Mesenchymal stem cells (MSCs) migrate via the bloodstream to sites of injury and are possibly attracted by inflammatory factors. As a proinflammatory mediator, angiotensin II (Ang II) reportedly enhances the migration of various cell types by signaling via the Ang II receptor in vitro. However, few studies have focused on the effects of Ang II on MSC migration and the underlying mechanisms. METHODS: Human bone marrow MSCs migration was measured using wound healing and Boyden chamber migration assays after treatments with different concentrations of Ang II, an AT1R antagonist (Losartan), and/or an AT2R antagonist (PD-123319). To exclude the effect of proliferation on MSC migration, we measured MSC proliferation after stimulation with the same concentration of Ang II. Additionally, we employed the focal adhesion kinase (FAK) inhibitor PF-573228, RhoA inhibitor C3 transferase, Rac1 inhibitor NSC23766, or Cdc42 inhibitor ML141 to investigate the role of cell adhesion proteins and the Rho-GTPase protein family (RhoA, Rac1, and Cdc42) in Ang II-mediated MSC migration. Cell adhesion proteins (FAK, Talin, and Vinculin) were detected by western blot analysis. The Rho-GTPase family protein activities were assessed by G-LISA and F-actin levels, which reflect actin cytoskeletal organization, were detected by using immunofluorescence. RESULTS: Human bone marrow MSCs constitutively expressed AT1R and AT2R. Additionally, Ang II increased MSC migration in an AT2R-dependent manner. Notably, Ang II-enhanced migration was not mediated by Ang II-mediated cell proliferation. Interestingly, Ang II-enhanced migration was mediated by FAK activation, which was critical for the formation of focal contacts, as evidenced by increased Talin and Vinculin expression. Moreover, RhoA and Cdc42 were activated by FAK to increase cytoskeletal organization, thus promoting cell contraction. Furthermore, FAK, Talin, and Vinculin activation and F-actin reorganization in response to Ang II were prevented by PD-123319 but not Losartan, indicating that FAK activation and F-actin reorganization were downstream of AT2R. CONCLUSIONS: These data indicate that Ang II-AT2R regulates human bone marrow MSC migration by signaling through the FAK and RhoA/Cdc42 pathways. This study provides insights into the mechanisms by which MSCs home to injury sites and will enable the rational design of targeted therapies to improve MSC engraftment.


Angiotensin II/pharmacology , Cell Movement/drug effects , Focal Adhesion Kinase 1/metabolism , Mesenchymal Stem Cells/metabolism , Receptor, Angiotensin, Type 2/metabolism , Signal Transduction/drug effects , cdc42 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Focal Adhesion Kinase 1/antagonists & inhibitors , Humans , Mesenchymal Stem Cells/cytology , rhoA GTP-Binding Protein/antagonists & inhibitors
19.
Crit Care Med ; 45(7): e727-e733, 2017 Jul.
Article En | MEDLINE | ID: mdl-28441237

OBJECTIVE: To evaluate the effectiveness of noninvasive ventilation in patients with acute hypoxemic nonhypercapnic respiratory failure unrelated to exacerbation of chronic obstructive pulmonary disease and cardiogenic pulmonary edema. DATA SOURCES: PubMed, EMBASE, Cochrane library, Web of Science, and bibliographies of articles were retrieved inception until June 2016. STUDY SELECTION: Randomized controlled trials comparing application of noninvasive ventilation with standard oxygen therapy in adults with acute hypoxemic nonhypercapnic respiratory failure were included. Chronic obstructive pulmonary disease exacerbation and cardiogenic pulmonary edema patients were excluded. The primary outcome was intubation rate; ICU mortality and hospital mortality were secondary outcomes. DATA EXTRACTION: Demographic variables, noninvasive ventilation application, and outcomes were retrieved. Internal validity was assessed using the risk of bias tool. The strength of evidence was assessed using Grading of Recommendations Assessment, Development, and Evaluation methodology. DATA SYNTHESIS: Eleven studies (1,480 patients) met the inclusion criteria and were analyzed by using a random effects model. Compared with standard oxygen therapy, the pooled effect showed that noninvasive ventilation significantly reduced intubation rate with a summary risk ratio of 0.59 (95% CI, 0.44-0.79; p = 0.0004). Furthermore, hospital mortality was also significantly reduced (risk ratio, 0.46; 95% CI, 0.24-0.87; p = 0.02). Subgroup meta-analysis showed that the application of bilevel positive support ventilation (bilevel positive airway pressure) was associated with a reduction in ICU mortality (p = 0.007). Helmet noninvasive ventilation could reduce hospital mortality (p = 0.0004), whereas face/nasal mask noninvasive ventilation could not. CONCLUSIONS: Noninvasive ventilation decreased endotracheal intubation rates and hospital mortality in acute hypoxemia nonhypercapnic respiratory failure excluding chronic obstructive pulmonary disease exacerbation and cardiogenic pulmonary edema patients. There is no sufficient scientific evidence to recommend bilevel positive airway pressure or helmet due to the limited number of trials available. Large rigorous randomized trials are needed to answer these questions definitely.


Noninvasive Ventilation/methods , Respiratory Insufficiency/therapy , Acute Disease , Adult , Aged , Female , Hospital Mortality , Humans , Intubation, Intratracheal/statistics & numerical data , Male , Middle Aged , Randomized Controlled Trials as Topic
20.
Mediators Inflamm ; 2016: 2347938, 2016.
Article En | MEDLINE | ID: mdl-27313398

Recently, mesenchymal stem cells (MSC) have been proved to be beneficial in acute respiratory distress syndrome (ARDS). Vascular endothelial growth factor (VEGF) is an important angiogenesis factor that MSC release. However, the precise role of VEGF-expressing character of MSC in the MSC treatment for ARDS remains obscure. Here, we firstly knocked down the gene VEGF in MSC (MSC-ShVEGF) with lentiviral transduction. Then we injected the MSC-ShVEGF to rats with lipopolysaccharide-induced acute lung injury (ALI) via the tail vein. Data showed that MSC transplantation significantly increased VEGF levels in the lung, reduced lung permeability, protected lung endothelium from apoptosis, facilitated VE-cadherin recovery, controlled inflammation, and attenuated lung injury. However, VEGF gene knockdown in MSC led to relatively insufficient VEGF expression in the injured lung and significantly diminished the therapeutic effects of MSC on ALI, suggesting an important role of VEGF-expressing behavior of MSC in the maintenance of VEGF in the lung and the MSC treatment for ALI. Hence, we conclude that MSC restores the lung permeability and attenuates lung injury in rats with ALI in part by maintaining a "sufficient" VEGF level in the lung and the VEGF-expressing character of MSC plays a positive role in the therapeutic effects of MSC on ARDS.


Acute Lung Injury/metabolism , Mesenchymal Stem Cells/metabolism , Vascular Endothelial Growth Factors/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/therapy , Animals , Cells, Cultured , Edema/metabolism , Edema/pathology , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/toxicity , Lung/metabolism , Lung/pathology , Male , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factors/genetics
...