Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Int J Mol Sci ; 22(23)2021 Nov 27.
Article in English | MEDLINE | ID: mdl-34884645

ABSTRACT

BACKGROUND: Ventilator-induced lung injury (VILI) frequently worsens acute respiratory distress syndrome (ARDS) severity. Human mesenchymal stem/stromal cells (MSCs) offer considerable therapeutic promise, but the key impediments of clinical translation stem from limitations due to cell source and availability, and concerns regarding the loss of efficacy following cryopreservation. These experiments compared the efficacy of umbilical-cord-derived MSCs (UC-MSCs), a readily available and homogenous tissue source, to the previously more widely utilised bone-marrow-derived MSCs (BM-MSCs). We assessed their capacity to limit inflammation, resolve injury and enhance repair in relevant lung mechanical stretch models, and the impact of cryopreservation on therapeutic efficacy. METHODS: In series 1, confluent alveolar epithelial layers were subjected to cyclic mechanical stretch (22% equibiaxial strain) and wound injury, and the potential of the secretome from BM- and UC-derived MSCs to attenuate epithelial inflammation and cell death, and enhance wound repair was determined. In series 2, anesthetized rats underwent VILI, and later received, in a randomised manner, 1 × 107 MSCs/kg intravenously, that were: (i) fresh BM-MSCs, (ii) fresh UC-MSCs or (iii) cryopreserved UC-MSCs. Control animals received a vehicle (PBS). The extent of the resolution of inflammation and injury, and repair was measured at 24 h. RESULTS: Conditioned medium from BM-MSCs and UC-MSCs comparably decreased stretch-induced pulmonary epithelial inflammation and cell death. BM-MSCs and UC-MSCs comparably enhanced wound resolution. In animals subjected to VILI, both fresh BM-MSCs and UC-MSCs enhanced injury resolution and repair, while cryopreserved UC-MSCs comparably retained their efficacy. CONCLUSIONS: Cryopreserved UC-MSCs can reduce stretch-induced inflammation and cell death, enhance wound resolution, and enhance injury resolution and repair following VILI. Cryopreserved UC-MSCs represent a more abundant, cost-efficient, less variable and equally efficacious source of therapeutic MSC product.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology , Ventilator-Induced Lung Injury/therapy , Animals , Cell Line, Tumor , Cells, Cultured , Cryopreservation/methods , Culture Media, Conditioned , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Rats , Rats, Sprague-Dawley , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/therapy , Umbilical Cord/metabolism , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/pathology
2.
Anesthesiology ; 129(3): 502-516, 2018 09.
Article in English | MEDLINE | ID: mdl-29979191

ABSTRACT

WHAT WE ALREADY KNOW ABOUT THIS TOPIC: WHAT THIS ARTICLE TELLS US THAT IS NEW: BACKGROUND:: Human mesenchymal stromal cells demonstrate promise for acute respiratory distress syndrome, but current studies use highly heterogenous cell populations. We hypothesized that a syndecan 2 (CD362)-expressing human mesenchymal stromal cell subpopulation would attenuate Escherichia coli-induced lung injury and enhance resolution after ventilator-induced lung injury. METHODS: In vitro studies determined whether CD362 human mesenchymal stromal cells could modulate pulmonary epithelial inflammation, wound healing, and macrophage phagocytosis. Two in vivo rodent studies determined whether CD362 human mesenchymal stromal cells attenuated Escherichia coli-induced lung injury (n = 10/group) and enhanced resolution of ventilation-induced injury (n = 10/group). RESULTS: CD362 human mesenchymal stromal cells attenuated cytokine-induced epithelial nuclear factor kappa B activation, increased epithelial wound closure, and increased macrophage phagocytosis in vitro. CD362 human mesenchymal stromal cells attenuated Escherichia coli-induced injury in rodents, improving arterial oxygenation (mean ± SD, 83 ± 9 vs. 60 ± 8 mmHg, P < 0.05), improving lung compliance (mean ± SD: 0.66 ± 0.08 vs. 0.53 ± 0.09 ml · cm H2O, P < 0.05), reducing bacterial load (median [interquartile range], 1,895 [100-3,300] vs. 8,195 [4,260-8,690] colony-forming units, P < 0.05), and decreasing structural injury compared with vehicle. CD362 human mesenchymal stromal cells were more effective than CD362 human mesenchymal stromal cells and comparable to heterogenous human mesenchymal stromal cells. CD362 human mesenchymal stromal cells enhanced resolution after ventilator-induced lung injury in rodents, restoring arterial oxygenation (mean ± SD: 113 ± 11 vs. 89 ± 11 mmHg, P < 0.05) and lung static compliance (mean ± SD: 0.74 ± 0.07 vs. 0.45 ± 0.07 ml · cm H2O, P < 0.05), resolving lung inflammation, and restoring histologic structure compared with vehicle. CD362 human mesenchymal stromal cells efficacy was at least comparable to heterogenous human mesenchymal stromal cells. CONCLUSIONS: A CD362 human mesenchymal stromal cell population decreased Escherichia coli-induced pneumonia severity and enhanced recovery after ventilator-induced lung injury.


Subject(s)
Acute Lung Injury/therapy , Escherichia coli Infections/therapy , Mesenchymal Stem Cell Transplantation/methods , Syndecan-2/biosynthesis , Ventilator-Induced Lung Injury/therapy , A549 Cells , Acute Lung Injury/etiology , Acute Lung Injury/microbiology , Animals , Bone Marrow/metabolism , Escherichia coli/isolation & purification , Escherichia coli Infections/metabolism , Humans , Male , Mesenchymal Stem Cells/metabolism , Rats , Rats, Sprague-Dawley , U937 Cells , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/microbiology
4.
Crit Care Med ; 45(2): e202-e212, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27861182

ABSTRACT

OBJECTIVE: Although mesenchymal stem/stromal cells represent a promising therapeutic strategy for acute respiratory distress syndrome, clinical translation faces challenges, including scarcity of bone marrow donors, and reliance on bovine serum during mesenchymal stem/stromal cell proliferation. We wished to compare mesenchymal stem/stromal cells from human umbilical cord, grown in xeno-free conditions, with mesenchymal stem/stromal cells from human bone marrow, in a rat model of Escherichia coli pneumonia. In addition, we wished to determine the potential for umbilical cord-mesenchymal stem/stromal cells to reduce E. coli-induced oxidant injury. DESIGN: Randomized animal study. SETTING: University research laboratory. SUBJECTS: Male Sprague-Dawley rats. INTERVENTIONS: Acute respiratory distress syndrome was induced in rats by intratracheal instillation of E. coli (1.5-2 × 10 CFU/kg). "Series 1" compared the effects of freshly thawed cryopreserved umbilical cord-mesenchymal stem/stromal cells with bone marrow-mesenchymal stem/stromal cells on physiologic indices of lung injury, cellular infiltration, and E. coli colony counts in bronchoalveolar lavage. "Series 2" examined the effects of cryopreserved umbilical cord-mesenchymal stem/stromal cells on survival, as well as measures of injury, inflammation and oxidant stress, including production of reactive oxidative species, reactive oxidative species scavenging by superoxide dismutase-1 and superoxide dismutase-2. MEASUREMENTS AND MAIN RESULTS: In "Series 1," animals subjected to E. coli pneumonia who received umbilical cord-mesenchymal stem/stromal cells had improvements in oxygenation, respiratory static compliance, and wet-to-dry ratios comparable to bone marrow-mesenchymal stem/stromal cell treatment. E. coli colony-forming units in bronchoalveolar lavage were reduced in both cell therapy groups, despite a reduction in bronchoalveolar lavage neutrophils. In series 2, umbilical cord-mesenchymal stem/stromal cells enhanced animal survival and decreased alveolar protein and proinflammatory cytokine concentrations, whereas increasing interleukin-10 concentrations. Umbilical cord-mesenchymal stem/stromal cell therapy decreased nicotinamide adenine dinucleotide phosphate-oxidase 2 and inducible nitric oxide synthase and enhanced lung concentrations of superoxide dismutase-2, thereby reducing lung tissue reactive oxidative species concentrations. CONCLUSIONS: Our results demonstrate that freshly thawed cryopreserved xeno-free human umbilical cord-mesenchymal stem/stromal cells reduce the severity of rodent E. coli-induced acute respiratory distress syndrome. Umbilical cord-mesenchymal stem/stromal cells, therefore, represent an attractive option for future clinical trials in acute respiratory distress syndrome.


Subject(s)
Lung Injury/prevention & control , Mesenchymal Stem Cell Transplantation , Respiratory Distress Syndrome/therapy , Animals , Culture Media, Serum-Free , Disease Models, Animal , Escherichia coli Infections/complications , Humans , Male , Mesenchymal Stem Cells/physiology , Oxidative Stress , Pneumonia, Bacterial/complications , Pneumonia, Bacterial/therapy , Rats , Rats, Sprague-Dawley , Respiratory Distress Syndrome/etiology , Umbilical Cord/cytology
5.
Intensive Care Med Exp ; 4(1): 8, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27001525

ABSTRACT

BACKGROUND: Hypercapnia, with its associated acidosis (HCA), is a consequence of respiratory failure and is also seen in critically ill patients managed with conventional "protective" ventilation strategies. Nuclear factor kappa-B (NF-κB), a pivotal transcription factor, is activated in the setting of injury and repair and is central to innate immunity. We have previously established that HCA protects against ventilation-induced lung injury in vivo, potentially via a mechanism involving inhibition of NF-κB signaling. We wished to further elucidate the role and mechanism of HCA-mediated inhibition of the NF-κB pathway in attenuating stretch-induced injury in vitro. METHODS: Initial experiments examined the effect of HCA on cyclic stretch-induced inflammation and injury in human bronchial and alveolar epithelial cells. Subsequent experiments examined the role of the canonical NF-κB pathway in mediating stretch-induced injury and the mechanism of action of HCA. The contribution of pH versus CO2 in mediating this effect of HCA was also examined. RESULTS: Pulmonary epithelial high cyclic stretch (22 % equibiaxial strain) activated NF-κB, enhanced interleukin-8 (IL-8) production, caused cell injury, and reduced cell survival. In contrast, physiologic stretch (10 % strain) did not activate inflammation or cause cell injury. HCA reduced cyclic mechanical stretch-induced NF-κB activation, attenuated IL-8 production, reduced injury, and enhanced survival, in bronchial and alveolar epithelial cells, following shorter (24 h) and longer (120 h) cyclic mechanical stretch. Pre-conditioning with HCA was less effective than when HCA was applied after commencement of cell stretch. HCA prevented the stretch-induced breakdown of the NF-κB cytosolic inhibitor IκBα, while IκBα overexpression "occluded" the effect of HCA. These effects were mediated by a pH-dependent mechanism rather than via CO2 per se. CONCLUSIONS: HCA attenuates adverse mechanical stretch-induced epithelial injury and death, via a pH-dependent mechanism that inhibits the canonical NF-κB activation by preventing IκBα breakdown.

6.
Crit Care Med ; 44(4): e207-17, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26584194

ABSTRACT

OBJECTIVE: Diverse effects of hypercapnic acidosis are mediated via inhibition of nuclear factor-κB, a pivotal transcription factor, in the setting of injury, inflammation, and repair, but the underlying mechanisms of action of hypercapnic acidosis on this pathway is unclear. We aim to examine the effect of hypercapnic acidosis on the nuclear factor-κB pathway in the setting of Escherichia coli-induced lung injury and characterize the underlying mechanisms in subsequent in vitro studies. DESIGN: In vivo animal study and subsequent in vitro studies. SETTING: University Research Laboratory. SUBJECTS: Adult male Sprague-Dawley rats and pulmonary epithelial cells. INTERVENTIONS: Following pulmonary IκBα-SuperRepressor transgene overexpression or sham and intratracheal E. coli inoculation, rats underwent 4 hours of mechanical ventilation under normocapnia or hypercapnic acidosis, and nuclear factor-κB activation, animal survival, lung injury, and cytokine profile were assessed. Subsequent in vitro studies examined the effect of hypercapnic acidosis on specific nuclear factor-κB canonical pathway kinases via overexpression of these components and in vitro kinase activity assays. The effect of hypercapnic acidosis on the p50/p65 nuclear factor-κB heterodimer was then assessed. MEASUREMENTS AND MAIN RESULTS: Hypercapnic acidosis and IκBα-SuperRepressor transgene overexpression reduced E. coli-induced lung inflammation and injury, decreased nuclear factor-κB activity, and increased animal survival. Hypercapnic acidosis inhibited canonical nuclear factor-κB signaling via reduced phosphorylative activation, reducing IκB kinase-ß activation and intrinsic activity, thereby decreasing IκBα degradation, and subsequent nuclear factor-κB translocation. Hypercapnic acidosis also directly reduced DNA binding of the nuclear factor-κB p65 subunit, although this effect was less marked. CONCLUSIONS: Hypercapnic acidosis reduced E. coli inflammation and lung injury in vivo and reduced nuclear factor-κB activation predominantly by inhibiting the activation and intrinsic activity of IκB kinase-ß.


Subject(s)
Acidosis, Respiratory/metabolism , Hypercapnia/metabolism , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Animals , Escherichia coli , I-kappa B Kinase/metabolism , Lung Injury/metabolism , Male , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , Rats , Rats, Sprague-Dawley , Respiration, Artificial , Respiratory Distress Syndrome/metabolism , Sepsis , Signal Transduction
7.
Curr Opin Crit Care ; 22(1): 14-20, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26645555

ABSTRACT

PURPOSE OF REVIEW: Acute respiratory distress syndrome (ARDS) is a devastating disease process with a 40% mortality rate, and for which there is no therapy. Stem cells are an exciting potential therapy for ARDS, and are currently the subject of intensive ongoing research efforts. We review data concerning the therapeutic promise of cell-based therapies for ARDS. RECENT FINDINGS: Recent experimental studies suggest that cell-based therapies, particularly mesenchymal stem/stromal cells (MSCs), endothelial progenitor cells, and embryonic or induced pluripotent stem cells all offer considerable promise for ARDS. Of these cell types, mesenchymal stromal cells offer the greatest potential for allogeneic therapy, given the large body of preclinical data supporting their use, and the advanced state of our understanding of their diverse mechanisms of action. Although other stem cells such as EPCs also have therapeutic potential, greater barriers exist, particularly the requirement for autologous EPC therapy. Other stem cells, such as ESCs and iPSCs, are at an earlier stage in the translational process, but offer the hope of directly replacing injured lung tissue. Ultimately, lung-derived stem cells may offer the greatest hope for lung diseases, given their homeostatic role in replacing and repairing damaged native lung tissues.MSCs are currently in early phase clinical trials, the results of which will be of critical importance to subsequent translational efforts for MSCs in ARDS. A number of translational challenges exist, including minimizing variability in cell batches, developing standard tests for cell potency, and producing large amounts of clinical-grade cells for use in patients. SUMMARY: Cell-based therapies, particularly MSCs, offer considerable promise for the treatment of ARDS. Overcoming translational challenges will be important to fully realizing their therapeutic potential for ARDS.


Subject(s)
Acute Lung Injury/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Respiratory Distress Syndrome/therapy , Acute Lung Injury/diagnosis , Acute Lung Injury/mortality , Animals , Clinical Trials as Topic , Disease Models, Animal , Graft Rejection , Graft Survival , Humans , Mesenchymal Stem Cell Transplantation/adverse effects , Prognosis , Respiratory Distress Syndrome/diagnosis , Respiratory Distress Syndrome/mortality , Risk Assessment , Severity of Illness Index , Survival Analysis , Treatment Outcome
8.
Intensive Care Med Exp ; 3(1): 29, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26472334

ABSTRACT

BACKGROUND: The potential for mesenchymal stem cells (MSCs) to reduce the severity of experimental lung injury has been established in several pre-clinical studies. We have recently demonstrated that MSCs, and MSC-secreted factors (secretome), enhance lung repair and regeneration at 48 h following ventilation-induced lung injury (VILI). We wished to determine the potential for MSC therapy to exert beneficial effects in the early recovery phase following VILI when ongoing injury coexists with processes of repair, and to compare the efficacy of MSC therapy to the use of the secretome alone. METHODS: Male Sprague-Dawley rats were anesthetized, oro-tracheally intubated, and subjected to high stretch mechanical ventilation until lung compliance had declined by 50 % of baseline. Animals were then weaned from mechanical ventilation, and anesthesia discontinued. Once awake and spontaneously ventilating, animals received an intravenous injection of either rodent MSCs (10 million/kg), MSC-conditioned medium, fibroblasts (10 million/kg), or vehicle. Thereafter, the animals were allowed to recover and the extent of lung injury/repair was determined after 4 h. RESULTS: Treatment with MSCs diminished injury and enhanced recovery following VILI to a greater extent compared to MSC-conditioned medium, with fibroblasts proving ineffective. MSCs, but not their conditioned medium, attenuated indices of lung injury including oxygenation, respiratory compliance, and lung edema. Total lung water as assessed by wet:dry ratio, bronchoalveolar lavage total inflammatory cell, neutrophil counts, and alveolar IL-6 concentrations were reduced in the animals that received MSC therapy. CONCLUSIONS: The immunomodulating and/or reparative effect of MSCs is evident early after VILI in this model. MSC-conditioned medium was not as effective as the cells themselves in diminishing injury and restoring lung structure and function.

9.
Thorax ; 70(7): 625-35, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25986435

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSCs) demonstrate considerable promise in preclinical acute respiratory distress syndrome models. We wished to determine the efficacy and mechanisms of action of human MSCs (hMSCs) in the setting of acute lung injury induced by prolonged Escherichia coli pneumonia in the rat. METHODS: Adult male Sprague Dawley rats underwent intratracheal instillation of E. coli bacteria in all experiments. In Series 1, animals were randomised to intravenous administration of: (1) vehicle (phosphate buffered saline (PBS), 300 µL); (2) 1×10(7) fibroblasts/kg; (3) 1×10(7) hMSCs/kg or (4) 2×10(7) hMSCs/kg. Series 2 determined the lowest effective hMSC dose. Series 3 compared the efficacy of intratracheal versus intravenous hMSC administration, while Series 4 examined the efficacy of cryopreserved hMSC. Series 5 examined the efficacy of the hMSC secretome. Parallel in vitro experiments further assessed the potential for hMSCs to secrete LL-37 and modulate macrophage phagocytosis. RESULTS: hMSC therapy reduced the severity of rodent E. coli pneumonia, improving survival, decreasing lung injury, reducing lung bacterial load and suppressing inflammation. Doses as low as 5×10(6) hMSCs/kg were effective. Intratracheal hMSC therapy was as effective as intravenous hMSC. Cryopreserved hMSCs were also effective, while the hMSC secretome was less effective in this model. hMSC therapy enhanced macrophage phagocytic capacity and increased lung and systemic concentrations of the antimicrobial peptide LL37. CONCLUSIONS: hMSC therapy decreased E. coli induced pneumonia injury and reduced lung bacterial burden, potentially via enhanced macrophage phagocytosis and increased alveolar LL-37 concentrations.


Subject(s)
Acute Lung Injury/prevention & control , Escherichia coli Infections/prevention & control , Mesenchymal Stem Cell Transplantation/methods , Pneumonia, Bacterial/prevention & control , Acute Lung Injury/immunology , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Animals , Bacterial Load , Cryopreservation , Disease Models, Animal , Escherichia coli/isolation & purification , Escherichia coli Infections/complications , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Humans , Infusions, Intravenous , Intubation, Intratracheal , Lung/microbiology , Male , Phagocytosis , Pneumonia, Bacterial/complications , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Rats, Sprague-Dawley , Transplantation, Heterologous
10.
Hum Gene Ther ; 26(1): 36-46, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25382145

ABSTRACT

Inhibition of the proinflammatory transcription factor NF-κB has previously been shown to attenuate the inflammatory response in tissue after injury. However, the feasibility and efficacy of aerosolized adeno-associated viral (AAV) vector-delivered transgenes to inhibit the NF-κB pathway are less clear. Initial studies optimized the AAV vector for delivery of transgenes to the pulmonary epithelium. The effect of repeated nebulization on the integrity and transduction efficacy of the AAV vector was then examined. Subsequent in vivo studies examined the efficacy of aerosolized rAAV2/6 overexpressing the NF-κB inhibitor IκBα in a rodent endotoxin-induced lung injury model. Initial in vitro investigations indicated that rAAV2/6 was the most effective vector to transduce the lung epithelium, and maintained its integrity and transduction efficacy after repeated nebulization. In our in vivo studies, animals that received aerosolized rAAV2/6-IκBα demonstrated a significant increase in total IκBα levels in lung tissue relative to null vector-treated animals. Aerosolized rAAV2/6-IκBα attenuated endotoxin-induced bronchoalveolar lavage-detected neutrophilia, interleukin-6 and cytokine-induced neutrophil chemoattractant-1 levels, as well as total protein content, and decreased histologic indices of injury. These results demonstrate that aerosolized AAV vectors encoding human IκBα significantly attenuate endotoxin-mediated lung injury and may be a potential therapeutic candidate in the treatment of acute lung injury.


Subject(s)
Acute Lung Injury/etiology , Acute Lung Injury/therapy , Dependovirus/genetics , Genetic Vectors/genetics , I-kappa B Proteins/genetics , Administration, Inhalation , Animals , Cytokines/metabolism , Dependovirus/classification , Disease Models, Animal , Endotoxins/adverse effects , Epithelial Cells/metabolism , Gene Expression , Genetic Vectors/administration & dosage , Lipopolysaccharides/adverse effects , Lipopolysaccharides/immunology , Lung/metabolism , Lung/pathology , Male , Rats , Respiratory Mucosa/metabolism , Serogroup , Transduction, Genetic , Transgenes
11.
Anesthesiology ; 122(2): 363-73, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25490744

ABSTRACT

BACKGROUND: Rodent mesenchymal stem/stromal cells (MSCs) enhance repair after ventilator-induced lung injury (VILI). We wished to determine the therapeutic potential of human MSCs (hMSCs) in repairing the rodent lung. METHODS: In series 1, anesthetized rats underwent VILI (series 1A, n = 8 to 9 per group) or protective ventilation (series 1B, n = 4 per group). After VILI, they were randomized to intravenous administration of (1) vehicle (phosphate-buffered saline); (2) fibroblasts (1 × 10 cells/kg); or (3) human MSCs (1 × 10 cells/kg) and the effect on restoration of lung function and structure assessed. In series 2, the efficacy of hMSC doses of 1, 2, 5, and 10 million/kg was examined (n = 8 per group). Series 3 compared the efficacy of both intratracheal and intraperitoneal hMSC administration to intravascular delivery (n = 5-10 per group). Series 4 examined the efficacy of delayed hMSC administration (n = 8 per group). RESULTS: Human MSC's enhanced lung repair, restoring oxygenation (131 ± 19 vs. 103 ± 11 vs. 95 ± 11 mmHg, P = 0.004) compared to vehicle or fibroblast therapy, respectively. hMSCs improved lung compliance, reducing alveolar edema, and restoring lung architecture. hMSCs attenuated lung inflammation, decreasing alveolar cellular infiltration, and decreasing cytokine-induced neutrophil chemoattractant-1 and interleukin-6 while increasing keratinocyte growth factor concentrations. The lowest effective hMSC dose was 2 × 10 hMSC/kg. Intraperitoneal hMSC delivery was less effective than intratracheal or intravenous hMSC. hMSCs enhanced lung repair when administered at later time points after VILI. CONCLUSIONS: hMSC therapy demonstrates therapeutic potential in enhancing recovery after VILI.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells , Ventilator-Induced Lung Injury/therapy , Animals , Fibroblasts , Humans , Lung/pathology , Male , Rats , Rats, Sprague-Dawley , Ventilator-Induced Lung Injury/pathology
12.
Crit Care ; 17(2): R82, 2013 Apr 27.
Article in English | MEDLINE | ID: mdl-23622108

ABSTRACT

INTRODUCTION: Nuclear factor (NF)-κB is central to the pathogenesis of inflammation in acute lung injury, but also to inflammation resolution and repair. We wished to determine whether overexpression of the NF-κB inhibitor IκBα could modulate the severity of acute and prolonged pneumonia-induced lung injury in a series of prospective randomized animal studies. METHODS: Adult male Sprague-Dawley rats were randomized to undergo intratracheal instillation of (a) 5 × 109 adenoassociated virus (AAV) vectors encoding the IκBα transgene (5 × 109 AAV-IκBα); (b) 1 × 10¹° AAV-IκBα; (c) 5 × 10¹° AAV-IκBα; or (d) vehicle alone. After intratracheal inoculation with Escherichia coli, the severity of the lung injury was measured in one series over a 4-hour period (acute pneumonia), and in a second series after 72 hours (prolonged pneumonia). Additional experiments examined the effects of IκBα and null-gene overexpression on E. coli-induced and sham pneumonia. RESULTS: In acute pneumonia, IκBα dose-dependently decreased lung injury, improving arterial oxygenation and lung static compliance, reducing alveolar protein leak and histologic injury, and decreasing alveolar IL-1ß concentrations. Benefit was maximal at the intermediate (1 × 10¹°) IκBα vector dose; however, efficacy was diminished at the higher (5 × 10¹°) IκBα vector dose. In contrast, IκBα worsened prolonged pneumonia-induced lung injury, increased lung bacterial load, decreased lung compliance, and delayed resolution of the acute inflammatory response. CONCLUSIONS: Inhibition of pulmonary NF-κB activity reduces early pneumonia-induced injury, but worsens injury and bacterial load during prolonged pneumonia.


Subject(s)
Escherichia coli Infections/drug therapy , Escherichia coli Infections/pathology , I-kappa B Proteins/administration & dosage , NF-kappa B/antagonists & inhibitors , Pneumonia, Bacterial/drug therapy , Pneumonia, Bacterial/pathology , Severity of Illness Index , Acute Disease , Animals , Escherichia coli Infections/metabolism , Genetic Vectors/administration & dosage , Male , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Pneumonia, Bacterial/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley
13.
Anesthesiology ; 118(4): 924-32, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23377221

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSCs) have been demonstrated to attenuate acute lung injury when delivered by intravenous or intratracheal routes. The authors aimed to determine the efficacy of and mechanism of action of intratracheal MSC therapy and to compare their efficacy in enhancing lung repair after ventilation-induced lung injury with intravenous MSC therapy. METHODS: : After induction of anesthesia, rats were orotracheally intubated and subjected to ventilation-induced lung injury (respiratory rate 18(-1) min, P insp 35 cm H2O,) to produce severe lung injury. After recovery, animals were randomized to receive: (1) no therapy, n = 4; (2) intratracheal vehicle (phosphate-buffered saline, 300 µl, n = 8); (3) intratracheal fibroblasts (4 × 10 cells, n = 8); (4) intratracheal MSCs (4 × 10(6) cells, n = 8); (5) intratracheal conditioned medium (300 µl, n = 8); or (6) intravenous MSCs (4 × 10(6) cells, n = 4). The extent of recovery after acute lung injury and the inflammatory response was assessed after 48 h. RESULTS: Intratracheal MSC therapy enhanced repair after ventilation-induced lung injury, improving arterial oxygenation (mean ± SD, 146 ± 3.9 vs. 110.8 ± 21.5 mmHg), restoring lung compliance (1.04 ± 0.11 vs. 0.83 ± 0.06 ml · cm H2O(-1)), reducing total lung water, and decreasing lung inflammation and histologic injury compared with control. Intratracheal MSC therapy attenuated alveolar tumor necrosis factor-α (130 ± 43 vs. 488 ± 211 pg · ml(-1)) and interleukin-6 concentrations (138 ± 18 vs. 260 ± 82 pg · ml(-1)). The efficacy of intratracheal MSCs was comparable with intravenous MSC therapy. Intratracheal MSCs seemed to act via a paracine mechanism, with conditioned MSC medium also enhancing lung repair after injury. CONCLUSIONS: Intratracheal MSC therapy enhanced recovery after ventilation-induced lung injury via a paracrine mechanism, and was as effective as intravenous MSC therapy.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Ventilator-Induced Lung Injury/surgery , Animals , Disease Models, Animal , Interleukin-6/blood , Intubation, Intratracheal , Lung/physiopathology , Lung Compliance , Oxygen/blood , Rats , Rats, Sprague-Dawley , Trachea , Treatment Outcome , Tumor Necrosis Factor-alpha/blood , Ventilator-Induced Lung Injury/blood
14.
Intensive Care Med ; 37(10): 1680-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21755396

ABSTRACT

PURPOSE: Superoxide is produced by activated neutrophils during the inflammatory response to stimuli such as endotoxin, can directly or indirectly injure host cells, and has been implicated in the pathogenesis of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). We wished to determine the potential for pulmonary overexpression of the extracellular isoform of superoxide dismutase (EC-SOD) to reduce the severity of endotoxin-induced lung injury. METHODS: Animals were randomly allocated to undergo intratracheal instillation of (1) surfactant alone (vehicle); (2) adeno-associated virus (AAV) vectors containing a null transgene (AAV-null); and (3) adeno-associated virus vectors containing the EC-SOD transgene (AAV-EC-SOD) and endotoxin was subsequently administered intratracheally. Two additional groups were randomized to receive (1) vehicle or (2) AAV-EC-SOD, and to undergo sham (vehicle) injury. The severity of the lung injury was assessed in all animals 24 h later. RESULTS: Endotoxin produced a severe lung injury compared to sham injury. The AAV vector encoding EC-SOD increased lung EC-SOD concentrations, and enhanced the antioxidant capacity of the lung. EC-SOD overexpression decreased the severity of endotoxin-induced ALI, reducing the decrement in systemic oxygenation and lung compliance, decreasing lung permeability and decreasing histologic injury. EC-SOD attenuated pulmonary inflammation, decreased bronchoalveolar lavage neutrophil counts, and reduced interleukin-6 and CINC-1 concentrations. The AAV vector itself did not contribute to inflammation or to lung injury. CONCLUSIONS: Pulmonary overexpression of EC-SOD protects the lung against endotoxin-induced ALI.


Subject(s)
Acute Lung Injury/enzymology , Acute Lung Injury/etiology , Endotoxins/physiology , Superoxide Dismutase/biosynthesis , Acute Lung Injury/prevention & control , Animals , Male , Rats , Rats, Sprague-Dawley
15.
Crit Care ; 15(3): 224, 2011.
Article in English | MEDLINE | ID: mdl-21699743

ABSTRACT

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) confer substantial morbidity and mortality, and have no specific therapy. The accessibility of the distal lung epithelium via the airway route, and the relatively transient nature of ALI/ARDS, suggest that the disease may be amenable to gene-based therapies. Ongoing advances in our understanding of the pathophysiology of ALI/ARDS have revealed multiple therapeutic targets for gene-based approaches. Strategies to enhance or restore lung epithelial and/or endothelial cell function, to strengthen lung defense mechanisms against injury, to speed clearance of infection and to enhance the repair process following ALI/ARDS have all demonstrated promise in preclinical models. Despite three decades of gene therapy research, however, the clinical potential for gene-based approaches to lung diseases including ALI/ARDS remains to be realized. Multiple barriers to effective pulmonary gene therapy exist, including the pulmonary architecture, pulmonary defense mechanisms against inhaled particles, the immunogenicity of viral vectors and the poor transfection efficiency of nonviral delivery methods. Deficits remain in our knowledge regarding the optimal molecular targets for gene-based approaches. Encouragingly, recent progress in overcoming these barriers offers hope for the successful translation of gene-based approaches for ALI/ARDS to the clinical setting.


Subject(s)
Acute Lung Injury/genetics , Acute Lung Injury/therapy , Genetic Therapy/trends , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/therapy , Animals , Gene Transfer Techniques/trends , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Time Factors
16.
Cancer Epidemiol Biomarkers Prev ; 20(1): 148-59, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21098650

ABSTRACT

BACKGROUND: Previously, we showed that gene suppression commonly occurs across chromosome 2q14.2 in colorectal cancer, through a process of long-range epigenetic silencing (LRES), involving a combination of DNA methylation and repressive histone modifications. We now investigate whether LRES also occurs in prostate cancer across this 4-Mb region and whether differential DNA methylation of 2q14.2 genes could provide a regional panel of prostate cancer biomarkers. METHODS: We used highly sensitive DNA methylation headloop PCR assays that can detect 10 to 25 pg of methylated DNA with a specificity of at least 1:1,000, and chromatin immunoprecipitation assays to investigate regional epigenetic remodeling across 2q14.2 in prostate cancer, in a cohort of 195 primary prostate tumors and 90 matched normal controls. RESULTS: Prostate cancer cells exhibit concordant deacetylation and methylation of histone H3 Lysine 9 (H3K9Ac and H3K9me2, respectively), and localized DNA hypermethylation of EN1, SCTR, and INHBB and corresponding loss of H3K27me3. EN1 and SCTR were frequently methylated (65% and 53%, respectively), whereas INHBB was less frequently methylated. CONCLUSIONS: Consistent with LRES in colorectal cancer, we found regional epigenetic remodeling across 2q14.2 in prostate cancer. Concordant methylation of EN1 and SCTR was able to differentiate cancer from normal (P < 0.0001) and improved the diagnostic specificity of GSTP1 methylation for prostate cancer detection by 26%. IMPACT: For the first time we show that DNA methylation of EN1 and SCTR promoters provide potential novel biomarkers for prostate cancer detection and in combination with GSTP1 methylation can add increased specificity and sensitivity to improve diagnostic potential.


Subject(s)
Biomarkers, Tumor/genetics , Chromosomes, Human, Pair 2 , DNA Methylation , Prostatic Neoplasms/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Glutathione S-Transferase pi/genetics , Homeodomain Proteins/genetics , Humans , Inhibin-beta Subunits/genetics , Male , Polymerase Chain Reaction/methods , Prognosis , Prostatic Neoplasms/diagnosis , Receptors, G-Protein-Coupled/genetics , Receptors, Gastrointestinal Hormone/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
17.
Mol Ther ; 16(5): 907-15, 2008 May.
Article in English | MEDLINE | ID: mdl-18388925

ABSTRACT

Synthetic vectors for cystic fibrosis (CF) gene therapy are required that efficiently and safely transfect airway epithelial cells, rather than alveolar epithelial cells or macrophages, and that are nonimmunogenic, thus allowing for repeated delivery. We have compared several vector systems against these criteria including GL67, polyethylenimine (PEI) 22 and 25 kd and two new, synthetic vector formulations, comprising a cationic, receptor-targeting peptide K(16)GACSERSMNFCG (E), and the cationic liposomes (L) DHDTMA/DOPE or DOSEP3/DOPE. The lipid and peptide formulations self assemble into receptor-targeted nanocomplexes (RTNs) LED-1 and LED-2, respectively, on mixing with plasmid (D). LED-1 transfected airway epithelium efficiently, while LED-2 and GL67 preferentially transfected alveolar cells. PEI transfected airway epithelial cells with high efficiency, but was more toxic to the mice than the other formulations. On repeat dosing, LED-1 was equally as effective as the single dose, while GL67 was 30% less effective and PEI 22 kd displayed a 90% reduction of efficiency on repeated delivery. LED-1 thus was the only formulation that fulfilled the criteria for a CF gene therapy vector while GL67 and LED-2 may be appropriate for other respiratory diseases. Opportunities for PEI depend on a solution to its toxicity problems. LED-1 formulations were stable to nebulization, the most appropriate delivery method for CF.


Subject(s)
Cystic Fibrosis/therapy , Gene Transfer Techniques , Genetic Therapy/methods , Nanotechnology/methods , Animals , Caspase 3/metabolism , Cations , Female , Genetic Therapy/instrumentation , Genetic Vectors , Humans , Lung/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Models, Biological
19.
J Biol Chem ; 280(43): 35807-14, 2005 Oct 28.
Article in English | MEDLINE | ID: mdl-16040608

ABSTRACT

Glucocorticoids have been shown to produce rapid nongenomic responses in airway epithelia. By using an intracellular pH (pH(i)) spectrofluorescence imaging system and the NH4Cl acid-loading technique, we have shown that the synthetic glucocorticoid,dexamethasone, accelerated intracellular pH recovery after an acid load in a human bronchial epithelial cell line (16HBE14o- cells). Exposure to NH4Cl (20 mm) elicited an intracellular acidification, followed by a pH(i) recovery. Inhibition of the Na+/H+ exchanger decreased the steady-state pH(i) and antagonized the dexamethasone stimulation of pH(i) regulation. The rapid effect of dexamethasone on pH(i) was neither affected by the inhibitor of transcription, cycloheximide, nor by the classical glucocorticoid and mineralocorticoid receptors antagonists, RU486 and spironolactone, respectively. The dexamethasone effect on pH(i) regulation was reduced by inhibitors of adenylate cyclase, cAMP-dependent protein kinase and mitogenactivated protein kinase (ERK1/2). By using a PepTag assay system and Western blotting, we have shown that dexamethasone stimulated cAMP-dependent protein kinase and mitogen-activated protein kinase activities. Taken together our results provide evidence for the rapid stimulation of Na+/H+ exchange activity by glucocorticoids in bronchial epithelial cells via a nongenomic mechanism involving cAMP-dependent protein kinase and mitogen-activated protein kinase ERK1/2 pathways.


Subject(s)
Bronchi/cytology , Dexamethasone/pharmacology , Epithelial Cells/cytology , Sodium-Hydrogen Exchangers/metabolism , Anti-Inflammatory Agents/pharmacology , Bronchi/pathology , Calcium/metabolism , Cell Line , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Glucocorticoids/metabolism , Humans , Hydrogen-Ion Concentration , MAP Kinase Signaling System , Mifepristone/pharmacology , Mineralocorticoids/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pertussis Toxin/pharmacology , Phosphorylation , Spectrometry, Fluorescence , Steroids/metabolism , Time Factors
20.
J Immunol ; 174(3): 1638-46, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15661927

ABSTRACT

Cystic fibrosis (CF) is a genetic disease characterized by severe neutrophil-dominated airway inflammation. An important cause of inflammation in CF is Pseudomonas aeruginosa infection. We have evaluated the importance of a number of P. aeruginosa components, namely lipopeptides, LPS, and unmethylated CpG DNA, as proinflammatory stimuli in CF by characterizing the expression and functional activity of their cognate receptors, TLR2/6 or TLR2/1, TLR4, and TLR9, respectively, in a human tracheal epithelial line, CFTE29o(-), which is homozygous for the DeltaF508 CF transmembrane conductance regulator mutation. We also characterized TLR expression and function in a non-CF airway epithelial cell line 16HBE14o(-). Using RT-PCR, we demonstrated TLR mRNA expression. TLR cell surface expression was assessed by fluorescence microscopy. Lipopeptides, LPS, and unmethylated CpG DNA induced IL-8 and IL-6 protein production in a time- and dose-dependent manner. The CF and non-CF cell lines were largely similar in their TLR expression and relative TLR responses. ICAM-1 expression was also up-regulated in CFTE29o(-) cells following stimulation with each agonist. CF bronchoalveolar lavage fluid, which contains LPS, bacterial DNA, and neutrophil elastase (a neutrophil-derived protease that can activate TLR4), up-regulated an NF-kappaB-linked reporter gene and increased IL-8 protein production in CFTE29o(-) cells. This effect was abrogated by expression of dominant-negative versions of MyD88 or Mal, key signal transducers for TLRs, thereby implicating them as potential anti-inflammatory agents for CF.


Subject(s)
Cystic Fibrosis/immunology , Cystic Fibrosis/pathology , Inflammation Mediators/physiology , Membrane Glycoproteins/physiology , Receptors, Cell Surface/physiology , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Adaptor Proteins, Signal Transducing , Antigens, Differentiation/genetics , Antigens, Differentiation/physiology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/pharmacology , Bronchoalveolar Lavage Fluid/immunology , Cell Line , CpG Islands/physiology , Cystic Fibrosis/microbiology , Humans , Inflammation Mediators/agonists , Intercellular Adhesion Molecule-1/biosynthesis , Interleukin-6/biosynthesis , Interleukin-8/antagonists & inhibitors , Interleukin-8/biosynthesis , Lipopeptides , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Lipoproteins/antagonists & inhibitors , Lipoproteins/pharmacology , Membrane Glycoproteins/agonists , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/pharmacology , Myeloid Differentiation Factor 88 , NF-kappa B/antagonists & inhibitors , NF-kappa B/biosynthesis , NF-kappa B/genetics , Oligodeoxyribonucleotides/pharmacology , Oligopeptides/antagonists & inhibitors , Oligopeptides/pharmacology , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Receptors, Cell Surface/agonists , Receptors, Cell Surface/biosynthesis , Receptors, Immunologic/genetics , Receptors, Immunologic/physiology , Receptors, Interleukin-1 , Respiratory Mucosa/metabolism , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptor 9 , Toll-Like Receptors , U937 Cells , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...