Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Stem Cells Transl Med ; 1(5): 359-72, 2012 May.
Article in English | MEDLINE | ID: mdl-23197815

ABSTRACT

We describe human chorionic mesenchymal stem cell (hCMSC) lines obtained from the chorion of human term placenta with high therapeutic potential in human organ pathology. hCMSCs propagated for more than 100 doublings without a decrease in telomere length and with no telomerase activity. Cells were highly positive for the embryonic stem cell markers OCT-4, NANOG, SSEA-3, and TRA-1-60. In vitro, cells could be differentiated into neuron-like cells (ectoderm), adipocytes, osteoblasts, endothelial-like cells (mesoderm), and hepatocytes (endoderm)-derivatives of all three germ layers. hCMSCs effectively facilitated repair of injured epithelium as demonstrated in an ex vivo-perfused human lung preparation injured by Escherichia coli endotoxin and in in vitro human lung epithelial cultures. We conclude that the chorion of human term placenta is an abundant source of multipotent stem cells that are promising candidates for cell-based therapies.


Subject(s)
Cell Differentiation , Chorion/cytology , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Neoplasms/therapy , Placenta/cytology , Stem Cell Transplantation , Animals , Blotting, Western , Cell Lineage , Cell Proliferation , Chorion/metabolism , Ectoderm/cytology , Ectoderm/metabolism , Endoderm/cytology , Endoderm/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Infant, Newborn , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Multipotent Stem Cells/metabolism , Placenta/metabolism , Pregnancy , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Telomerase/metabolism , Telomere/genetics
2.
Thorax ; 67(6): 533-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22250097

ABSTRACT

RATIONALE: Bacterial pneumonia is the most common infectious cause of death worldwide and treatment is increasingly hampered by antibiotic resistance. Mesenchymal stem cells (MSCs) have been demonstrated to provide protection against acute inflammatory lung injury; however, their potential therapeutic role in the setting of bacterial pneumonia has not been well studied. OBJECTIVE: This study focused on testing the therapeutic and mechanistic effects of MSCs in a mouse model of Gram-negative pneumonia. METHODS AND RESULTS: Syngeneic MSCs from wild-type mice were isolated and administered via the intratracheal route to mice 4 h after the mice were infected with Escherichia coli. 3T3 fibroblasts and phosphate-buffered saline (PBS) were used as controls for all in vivo experiments. Survival, lung injury, bacterial counts and indices of inflammation were measured in each treatment group. Treatment with wild-type MSCs improved 48 h survival (MSC, 55%; 3T3, 8%; PBS, 0%; p<0.05 for MSC vs 3T3 and PBS groups) and lung injury compared with control mice. In addition, wild-type MSCs enhanced bacterial clearance from the alveolar space as early as 4 h after administration, an effect that was not observed with the other treatment groups. The antibacterial effect with MSCs was due, in part, to their upregulation of the antibacterial protein lipocalin 2. CONCLUSIONS: Treatment with MSCs enhanced survival and bacterial clearance in a mouse model of Gram-negative pneumonia. The bacterial clearance effect was due, in part, to the upregulation of lipocalin 2 production by MSCs.


Subject(s)
Acute-Phase Proteins/metabolism , Escherichia coli Infections/complications , Escherichia coli Infections/surgery , Escherichia coli/pathogenicity , Lipocalins/metabolism , Mesenchymal Stem Cell Transplantation , Oncogene Proteins/metabolism , Pneumonia, Bacterial/microbiology , Acute-Phase Proteins/biosynthesis , Animals , Disease Models, Animal , Lipocalin-2 , Lipocalins/biosynthesis , Mesenchymal Stem Cell Transplantation/methods , Mice , Mice, Inbred C57BL , Oncogene Proteins/biosynthesis , Pneumonia, Bacterial/surgery , Survival Analysis , Trachea , Treatment Outcome , Up-Regulation
3.
Stem Cells Dev ; 21(4): 589-97, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21631154

ABSTRACT

Proteins p130 and E2f4, members of the retinoblastoma protein (pRb) family/E2F transcription factor family, are the key elements in regulation of cell cycle and differentiation. The functional role of the p130/E2f4 in mesenchymal stem cells (MSC) is unclear. We demonstrate here that activation of the Wnt/ß-catenin pathway in mouse MSC is associated with accumulation of active forms of the p130, E2f4, and ß-catenin but does not result in inhibition of cell cycle progression. The levels and phosphorylation patterns of p130, E2f4, and ß-catenin in MSC do not change during cell cycle progression. This is different from control T98G glyoblastoma cells that accumulated differently phosphorylated forms of the p130 in quiescence, and under active proliferation. In MSC, synchronized at G0/G1 and S cell cycle phases, the p130 and ß-catenin physically interact each other, whereas Gsk3ß was associated and co-precipitated with both p130 and ß-catenin. Our results indicate that Wnt/ß-catenin and pRb signal pathways interact with each other and form common p130/Gsk3ß/ß-catenin complex during MSC cycle progression. Physiological relevance of such complex may be associated with coupling of the cell cycle and differentiation in MSC, which is related to a wide differentiation potential of these stem cells.


Subject(s)
E2F4 Transcription Factor/metabolism , Glycogen Synthase Kinase 3/metabolism , Mesenchymal Stem Cells/metabolism , Retinoblastoma-Like Protein p130/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Cell Cycle/physiology , Cell Line, Tumor , E2F4 Transcription Factor/genetics , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Transgenic , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phosphorylation/physiology , Retinoblastoma-Like Protein p130/genetics , Wnt Proteins/genetics , beta Catenin/genetics
4.
Stem Cells ; 28(12): 2229-38, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20945332

ABSTRACT

Recent in vivo studies indicate that mesenchymal stem cells (MSCs) may have beneficial effects in the treatment of sepsis induced by bacterial infection. Administration of MSCs in these studies improved survival and enhanced bacterial clearance. The primary objective of this study was to test the hypothesis that human MSCs possessed intrinsic antimicrobial properties. We studied the effect of human MSCs derived from bone marrow on the bacterial growth of Gram-negative (Escherichia coli and Pseudomonas aeruginosa) and Gram-positive (Staphylococcus aureus) bacteria. MSCs as well as their conditioned medium (CM) demonstrated marked inhibition of bacterial growth in comparison with control medium or normal human lung fibroblasts (NHLF). Analysis of expression of major antimicrobial peptides indicated that one of the factors responsible for the antimicrobial activity of MSC CM against Gram-negative bacteria was the human cathelicidin antimicrobial peptide, hCAP-18/LL-37. Both m-RNA and protein expression data showed that the expression of LL-37 in MSCs increased after bacterial challenge. Using an in vivo mouse model of E. coli pneumonia, intratracheal administration of MSCs reduced bacterial growth (in colony-forming unit) in the lung homogenates and in the bronchoalveolar lavage (BAL) fluid, and administration of MSCs simultaneously with a neutralizing antibody to LL-37 resulted in a decrease in bacterial clearance. In addition, the BAL itself from MSC-treated mice had a greater antimicrobial activity in comparison with the BAL of phosphate buffered saline (PBS)-treated mice. Human bone marrow-derived MSCs possess direct antimicrobial activity, which is mediated in part by the secretion of human cathelicidin hCAP-18/ LL-37.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cathelicidins/metabolism , Cathelicidins/pharmacology , Mesenchymal Stem Cells/metabolism , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Animals , Anti-Bacterial Agents/therapeutic use , Antibodies/administration & dosage , Antibodies/pharmacology , Antimicrobial Cationic Peptides , Bronchoalveolar Lavage Fluid/cytology , Cathelicidins/therapeutic use , Cell Count , Coculture Techniques , Culture Media, Conditioned/pharmacology , Escherichia coli/cytology , Escherichia coli/drug effects , Escherichia coli/growth & development , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Neutralization Tests , Pneumonia/drug therapy , Pneumonia/microbiology , Up-Regulation/drug effects
5.
Proc Natl Acad Sci U S A ; 106(38): 16357-62, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19721001

ABSTRACT

Recent studies have suggested that bone marrow-derived multipotent mesenchymal stem cells (MSCs) may have therapeutic applications in multiple clinical disorders including myocardial infarction, diabetes, sepsis, and hepatic and acute renal failure. Here, we tested the therapeutic capacity of human MSCs to restore alveolar epithelial fluid transport and lung fluid balance from acute lung injury (ALI) in an ex vivo perfused human lung preparation injured by E. coli endotoxin. Intra-bronchial instillation of endotoxin into the distal airspaces resulted in pulmonary edema with the loss of alveolar epithelial fluid transport measured as alveolar fluid clearance. Treatment with allogeneic human MSCs or its conditioned medium given 1 h following endotoxin-induced lung injury reduced extravascular lung water, improved lung endothelial barrier permeability and restored alveolar fluid clearance. Using siRNA knockdown of potential paracrine soluble factors, secretion of keratinocyte growth factor was essential for the beneficial effect of MSCs on alveolar epithelial fluid transport, in part by restoring amiloride-dependent sodium transport. In summary, treatment with allogeneic human MSCs or the conditioned medium restores normal fluid balance in an ex vivo perfused human lung injured by E. coli endotoxin.


Subject(s)
Lung Injury/surgery , Lung/surgery , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Acute Disease , Adult , Amiloride/pharmacology , Body Fluids/metabolism , Cells, Cultured , Culture Media, Conditioned/pharmacology , Endotoxins , Escherichia coli/chemistry , Female , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/pharmacology , Humans , In Vitro Techniques , Leukocyte Count , Lung/metabolism , Lung/pathology , Lung Injury/chemically induced , Male , Mesenchymal Stem Cells/cytology , Middle Aged , Neutrophils/pathology , Perfusion , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Pulmonary Edema/prevention & control , Recombinant Proteins/pharmacology , Sodium Channel Blockers/pharmacology , Transplantation, Homologous
6.
Expert Opin Biol Ther ; 9(10): 1259-70, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19691441

ABSTRACT

Despite extensive research into the pathogenesis of acute lung injury and the acute respiratory distress syndrome (ALI/ARDS), mortality remains high at approximately 40%. Current treatment is primarily supportive, with lung-protective ventilation and a fluid conservative strategy. Pharmacologic therapies that reduce the severity of lung injury in experimental studies have not yet been translated into effective clinical treatment options. Therefore, innovative therapies are needed. Recent studies have suggested that bone-marrow-derived multipotent mesenchymal stem cells (MSC) may have therapeutic applications in multiple clinical disorders including myocardial infarction, diabetes, sepsis, hepatic and acute renal failure. Recently, MSC have been studied in several in vivo models of lung disease. This review focuses on first describing the existing experimental literature that has tested the use of MSC in models of ALI/ARDS, and then the potential mechanisms underlying their therapeutic use with an emphasis on secreted paracrine soluble factors. The review concludes with a discussion of future research directions required for potential clinical trials.


Subject(s)
Acute Lung Injury/surgery , Mesenchymal Stem Cells/cytology , Stem Cell Transplantation , Acute Lung Injury/physiopathology , Animals , Humans
7.
Exp Biol Med (Maywood) ; 234(7): 813-23, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19429852

ABSTRACT

The main barrier to a broader clinical application of umbilical cord blood (UCB) transplantation is its limiting cellular content. Thus, the discovery of hematopoietic progenitor cells in murine placental tissue led us investigate whether the human placenta contains hematopoietic cells, sites of hematopoiesis, and to develop a procedure of processing and storing placental hematopoietic cells for transplantation. Here we show that the human placenta contains large numbers of CD34-expressing hematopoietic cells, with the potential to provide a cellular yield several-fold greater than that of a typical UCB harvest. Cells from fresh or cryopreserved placental tissue generated erythroid and myeloid colonies in culture, and also produced lymphoid cells after transplantation in immunodeficient mice. These results suggest that human placenta could become an important new source of hematopoietic cells for allogeneic transplantation.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Placenta/cytology , Animals , Antigens, CD34/metabolism , Cell Differentiation/physiology , Cell Separation/methods , Cells, Cultured , Cryopreservation , Female , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Humans , Mice , Mice, Inbred NOD , Pregnancy
8.
Lung ; 186(3): 179-190, 2008.
Article in English | MEDLINE | ID: mdl-18357492

ABSTRACT

Bone marrow-derived cells (BMDC) have been shown to graft injured tissues, differentiate in specialized cells, and participate in repair. The importance of these processes in acute lung bacterial inflammation and development of fibrosis is unknown. The goal of this study was to investigate the temporal sequence and lineage commitment of BMDC in mouse lungs injured by bacterial pneumonia. We transplanted GFP-tagged BMDC into 5-Gy-irradiated C57BL/6 mice. After 3 months of recovery, mice were subjected to LD(50) intratracheal instillation of live E. coli (controls received saline) which produced pneumonia and subsequent areas of fibrosis. Lungs were investigated by immunohistology for up to 6 months. At the peak of lung inflammation, the predominant influx of BMDC were GFP(+) leukocytes. Postinflammatory foci of lung fibrosis were evident after 1-2 months. The fibrotic foci in lung stroma contained clusters of GFP(+) CD45(+) cells, GFP(+) vimentin-positive cells, and GFP(+) collagen I-positive fibroblasts. GFP(+) endothelial or epithelial cells were not identified. These data suggest that following 5-Gy irradiation and acute bacterial pneumonia, BMDC may temporarily participate in lung postinflammatory repair and stromal remodeling without long-term engraftment as specialized endothelial or epithelial cells.


Subject(s)
Bone Marrow Cells/pathology , Lung/pathology , Macrophages/pathology , Pneumonia, Bacterial/pathology , Stem Cells/pathology , Acute Disease , Animals , Bone Marrow Cells/immunology , Disease Models, Animal , Fluorescent Antibody Technique , Immunity, Cellular , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Stem Cells/immunology
9.
J Lipid Res ; 49(5): 1103-12, 2008 May.
Article in English | MEDLINE | ID: mdl-18268358

ABSTRACT

Human acyl-coenzyme A binding domain-containing member 6 (ACBD6) is a modular protein that carries an acyl-CoA binding domain at its N terminus and two ankyrin motifs at its C terminus. ACBD6 binds long-chain acyl-CoAs with a strong preference for unsaturated, C18:1-CoA and C20:4-CoA, over saturated, C16:0-CoA, acyl species. Deletion of the C terminus, which is not conserved among the members of this family, did not affect the binding capacity or the substrate specificity of the protein. ACBD6 is not a ubiquitous protein, and its expression is restricted to tissues and progenitor cells with functions in blood and vessel development. ACBD6 was detected in bone marrow, spleen, placenta, cord blood, circulating CD34+ progenitors, and embryonic-like stem cells derived from placenta. In placenta, the protein was only detected in CD34+ progenitor cells present in blood and in CD31+ endothelial cells surrounding the blood vessels. These cells were also positive for the marker CD133, and they probably constitute hemangiogenic stem cells, precursors of both blood and vessels. We propose that human ACBD6 represents a cellular marker for primitive progenitor cells with functions in hematopoiesis and vascular endothelium development.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Binding Sites , Chromatography, Gel , DNA/genetics , Embryonic Stem Cells/physiology , Female , Humans , Placenta/physiology , Pregnancy , RNA/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spleen/physiology
10.
Lung ; 186(2): 103-14, 2008.
Article in English | MEDLINE | ID: mdl-18219526

ABSTRACT

The aim of this study was to determine the functional significance of peroxiredoxin V (PRXV) in defense against oxidative stress and changes of its expression in human lung inflammation. We used in vitro cell cultures and retrospective analyses of human sputum samples to perform the study. We found that stable clones of lung epithelial cell lines A549 and U1810 with reduced expression of PRXV were prone to oxidative damage. Upregulation of PRXV decreased induction of DNA double-strand breaks and protein oxidation by cigarette smoke extract and hydrogen peroxide. Transfection with PRXV-carrying plasmid protected Calu-3 confluent epithelial cell sheets from alterations in barrier permeability induced by oxidative stress. In human sputum proinflammatory cytokines, myeloperoxidase, and PRXV were increased during viral-induced inflammation. We conclude that PRXV is an important antioxidant protein of lung epithelial cells. Its expression in the human lung increases in inflammation.


Subject(s)
Antioxidants/metabolism , Epithelial Cells/metabolism , Lung/metabolism , Peroxiredoxins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Common Cold/metabolism , Cytokines/metabolism , Epithelial Cells/pathology , Humans , Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Oxidative Stress/physiology , Retrospective Studies , Sputum/metabolism , Transfection
11.
FEBS J ; 274(22): 5804-14, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17937766

ABSTRACT

Peroxiredoxin V (PRDX5) is a member of the family of mammalian proteins that neutralize reactive oxygen species. The PRDX5 gene is constitutively expressed at a high level in many human tissues, but functional elements of its promoter responsible for a high basal activity in the absence of oxidative stress have still not been identified. Among predicted binding sites for transcription factors in the human PRDX5 promoter are binding sites for nuclear respiratory factor 1 (NFR-1) and nuclear respiratory factor 2 (also called GABPA), which regulate the biogenesis of mitochondria. We constructed luciferase reporter gene plasmids containing stepwise deletions of the PRDX5 promoter and examined their activities in transient transfections. Our results suggest that basal PRDX5 promoter activity mostly depends on NFR-1 and GABPA sites. The latter, in the PRDX5 promoter, were conserved in the six mammalian genomes analyzed (human, chimpanzee, cow, mouse, rat and dog) and a fraction of human PRDX5 associates with the mitochondrial matrix. We also found that the N-terminal 50 amino acids of the full-length human PRDX5 (24 kDa) translated from its first AUG codon targets this protein exclusively to mitochondria. However, the short form of PRDX5 (17 kDa), translated from its second AUG codon, has cytoplasmic and nuclear localization, which is also typical for endogenously expressed protein. Together, our results indicate that high basal expression of the PRDX5 gene is coordinated with the expression of nuclear genes encoding mitochondrial proteins and that the PRDX5 protein might play a major role in permanent defense against reactive oxygen species produced by mitochondria.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation/physiology , Mitochondria/metabolism , Peroxiredoxins/metabolism , Transcription Factors/physiology , Base Sequence , Cell Line , DNA Primers , Humans , Microscopy, Fluorescence , Peroxiredoxins/genetics , Promoter Regions, Genetic
12.
Anat Rec (Hoboken) ; 290(9): 1033-45, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17661377

ABSTRACT

The goal of the study was to investigate participation of bone marrow (BM) cells in the process of airway epithelial restoration after naphthalene-induced injury. We transplanted sex-mismatched green fluorescent protein (GFP) -tagged BM-derived cultured plastic-adherent mesenchymal stem cells into 5Gy-irradiated C57BL/6 recipients. After 1 month of recovery, experimental animals were subjected to 250 mg/kg naphthalene IP. Animals were killed at 2-30 days after naphthalene. By immunofluorescence, immunohistochemistry, and by in situ hybridization for the Y-chromosome, we observed patches of donor-derived cells in the large and small conducting airways, mostly at 2-6 days after injury. GFP(+) cells in the epithelium of airways were positive for pancytokeratin and some other epithelial markers. Although rare, GFP(+) cells formed clear isolated patches of the bronchial epithelium, consistent with clonal formation; as some cells were also positive for proliferating cell nuclear antigen, a marker of proliferating cells. After day 12, only occasional GFP(+) cells were present in the epithelium. These data confirm that bone marrow-derived cultured mesenchymal cells can participate in the recovery of the injured airway epithelium after naphthalene-induced injury with minimal long-term engraftment.


Subject(s)
Bone Marrow Cells/physiology , Cell Proliferation , Lung Diseases/physiopathology , Lung/physiopathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Regeneration , Animals , Bone Marrow Cells/metabolism , Cells, Cultured , Clone Cells/physiology , Disease Models, Animal , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lung/pathology , Lung/surgery , Lung Diseases/chemically induced , Lung Diseases/pathology , Lung Diseases/surgery , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Naphthalenes , Phenotype , Proliferating Cell Nuclear Antigen/metabolism , Respiratory Mucosa/pathology , Respiratory Mucosa/physiopathology , Time Factors , Whole-Body Irradiation
13.
J Immunol ; 179(3): 1855-63, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17641052

ABSTRACT

Recent in vivo and in vitro work suggests that mesenchymal stem cells (MSC) have anti-inflammatory properties. In this study, we tested the effect of administering MSC directly into the airspaces of the lung 4 h after the intrapulmonary administration of Escherichia coli endotoxin (5 mg/kg). MSC increased survival compared with PBS-treated control mice at 48 h (80 vs 42%; p < 0.01). There was also a significant decrease in excess lung water, a measure of pulmonary edema (145 +/- 50 vs 87 +/- 20 microl; p < 0.01), and bronchoalveolar lavage protein, a measure of endothelial and alveolar epithelial permeability (3.1 +/- 0.4 vs 2.2 +/- 0.8 mg/ml; p < 0.01), in the MSC-treated mice. These protective effects were not replicated by the use of further controls including fibroblasts and apoptotic MSC. The beneficial effect of MSC was independent of the ability of the cells to engraft in the lung and was not related to clearance of the endotoxin by the MSC. MSC administration mediated a down-regulation of proinflammatory responses to endotoxin (reducing TNF-alpha and MIP-2 in the bronchoalveolar lavage and plasma) while increasing the anti-inflammatory cytokine IL-10. In vitro coculture studies of MSC with alveolar macrophages provided evidence that the anti-inflammatory effect was paracrine and was not cell contact dependent. In conclusion, treatment with intrapulmonary MSC markedly decreases the severity of endotoxin-induced acute lung injury and improves survival in mice.


Subject(s)
Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/pathology , Lipopolysaccharides/toxicity , Lung/immunology , Lung/pathology , Mesenchymal Stem Cell Transplantation , 3T3 Cells , Animals , Bone Marrow Transplantation/mortality , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Cell Differentiation , Cells, Cultured , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Cytokines/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Intubation, Intratracheal , Lung/metabolism , Male , Mesenchymal Stem Cell Transplantation/mortality , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL
14.
Tissue Eng ; 13(10): 2441-50, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17630877

ABSTRACT

Mesenchymal stem cells (MSCs) from bone marrow are a potential source for reconstructive therapy. In vitro, MSCs differentiate into cells of mesodermal and ectodermal lineages but rarely into cells of endodermal lineage. We developed an in vitro model to study the endodermal differentiation of MSCs using co-culture of MSCs and transformed lung epithelial (A-549) cells. The cells were separated using a cell-impermeable membrane to eliminate the possibility of cell fusion. Under these conditions, MSCs expressed several lung epithelial markers (cytokeratins 5, 8, 14, 18, 19, pro-surfactant protein C, zonula occludens-1), detected using quantitative reverse transcriptase polymerase chain reaction and Western blot, and beta-catenin signaling was activated in MSCs. Treatment of MSCs with 10 to 20 mM lithium chloride activated the beta-catenin pathway and enhanced expression of epithelial markers, although this activation was transient. We conclude that A-549 cells can trigger epithelial differentiation of MSCs by a paracrine mechanism that may include activation of beta-catenin signaling.


Subject(s)
Bone Marrow Cells/cytology , Coculture Techniques/methods , Endoderm/cytology , Epithelial Cells/cytology , Lung/cytology , Mesenchymal Stem Cells/cytology , Paracrine Communication/physiology , Animals , Bone Marrow Cells/physiology , Cell Differentiation , Cells, Cultured , Endoderm/physiology , Epithelial Cells/physiology , Mesenchymal Stem Cells/physiology , Mice
15.
Immunol Invest ; 36(1): 59-72, 2007.
Article in English | MEDLINE | ID: mdl-17190650

ABSTRACT

The role of ICAM-1 in contact activation of the bronchial epithelial cells is elucidated. Direct contact between epithelial cells and leukocytes is required to change transepithelial electrical resistance (TER) of the epithelium. Migration of human neutrophils across the layers of cultured human airway epithelial cells (Calu-3) or primary cow tracheal epithelial cells was induced by an fMLP gradient. Migrating neutrophils decreased TER and increased permeability to albumin. Monoclonal antibodies to ICAM-1 reduced neutrophil migration, thus reducing the changes in TER and changes in the epithelial permeability to albumin. By confocal microscopy, ERK1/2 was found to be locally activated in the epithelial cells at the sites of migration and cross-linking of ICAM-1. Blockade of ERK1/2 by PD98059 decreased the changes in TER which were induced by ICAM-1 cross-linking. Contact activation of the bronchial epithelial cells, involving ICAM-1 via local activation of ERK1/2, is an important mechanism of alteration of the bronchial epithelial permeability.


Subject(s)
Epithelial Cells/physiology , Intercellular Adhesion Molecule-1/metabolism , Lung/physiology , Trachea/cytology , Trachea/physiology , Albumins/metabolism , Animals , Antibodies/immunology , Cattle , Cell Movement , Cells, Cultured , Enzyme Activation , Humans , Intercellular Adhesion Molecule-1/immunology , Leukocytes/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Permeability , Phosphorylation , Tumor Cells, Cultured
16.
J Inflamm (Lond) ; 3: 13, 2006 Oct 04.
Article in English | MEDLINE | ID: mdl-17020618

ABSTRACT

BACKGROUND: We characterized changes in expression of the antioxidant protein Peroxiredoxin V (PRXV) during airway inflammation. METHODS: Studies in anesthetized rats and mice; PRXV expression determined by Western blot analyses and immunohistochemistry; PRXV m-RNA expression determined by Taq-Man RT-PCR. RESULTS: Bacterial lung inflammation did not change expression of PRXV in murine epithelia but produced massive influx of leukocytes highly expressing PRXV. Endotoxin and f-MLP induced leukocyte migration in rat trachea but did not change mRNA levels and PRXV protein expression in tracheal epithelial cells. In primary airway cell culture (cow), alveolar epithelial cells A549, or co-culture of A549 with murine macrophages RAW264.7, exposure to live bacteria increased expression of PRXV, which required serum. PRXV was secreted in vitro by epithelial and immune cells. CONCLUSION: Inflammation increased expression of PRXV in airways by at least 2 mechanisms: cell population shift by massive influx of leukocytes expressing PRXV, and moderate post-transcriptional up-regulation of PRXV in epithelial cells.

17.
FEBS J ; 273(12): 2607-17, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16817890

ABSTRACT

Peroxiredoxins belong to a family of antioxidant proteins that neutralize reactive oxygen species. One member of this family, peroxiredoxin I (PRDX1), suppresses DNA oxidation. Peroxiredoxin V (PRDX5) has been cloned as a transcriptional corepressor, as a peroxisomal/mitochondrial antioxidant protein, and as an inhibitor of p53-dependent apoptosis. Promoters of mammalian PRDX5 genes contain clusters of antioxidant response elements, which can bind the transcription factor NRF2. However, we found that expression of the human PRDX5 gene in situ was not stimulated by the oxidative agent menadione. Silencing of the NRF2 gene in the absence of oxidative stress by specific siRNA did not decrease PRDX5 protein concentration. We also constructed clones of human lung epithelial cells A549 with siRNA-mediated knockdown of the PRDX5 gene. This led to a significant increase in 8-oxoguanine formation in cell DNA. In the PRDX5 knockdown clone, an increase in transcripts containing sequences of alpha-satellite and satellite III DNAs was also detected, suggesting that this protein may be required for silencing of heterochromatin. Together, these results suggest that constitutively expressed PRDX5 gene plays an important role in protecting the genome against oxidation and may also be involved in the control of transcription of noncoding DNA.


Subject(s)
DNA Damage , DNA, Satellite/genetics , Gene Expression Regulation , Oxidative Stress , Peroxidases/genetics , Peroxidases/physiology , Base Sequence , Cloning, Molecular , Guanosine/analogs & derivatives , Guanosine/metabolism , Humans , Models, Genetic , Molecular Sequence Data , NF-E2-Related Factor 2/genetics , Oxidation-Reduction , Oxidative Stress/genetics , Peroxidases/metabolism , Peroxiredoxins , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Tumor Cells, Cultured
18.
Exp Cell Res ; 312(15): 2806-15, 2006 Sep 10.
Article in English | MEDLINE | ID: mdl-16781710

ABSTRACT

Sensitivity of tumor cells to treatment with anticancer drugs depends on expression and function of antiapoptotic and antioxidant proteins. The goal of our study was to determine the functional role of the novel antioxidant protein Peroxiredoxin V (PrxV), in protection of human lung carcinoma cell lines against apoptosis. Analysis of expression of PrxV in multiple lung carcinoma cell lines revealed a positive correlation between the expression of PrxV and radioresistance in vitro. Clones of the lung carcinoma cells U1810 with down-regulated expression of PrxV, or with its impaired enzymatic function (expression of redox-negative PrxV), demonstrated increased sensitivity to treatment with anticancer drugs etoposide and adriamycin. Pre-treatment of these clones with antioxidant N-acetyl-cysteine did not change their sensitivity to adriamycin, suggesting the involvement of a non-redox activity of PrxV. Expression of the redox-negative PrxV mainly affected the mitochondrial pathway of apoptosis, as assessed by cytochrome c release assay. Impairment of the PrxV enzymatic function also affected transmembrane potential and calcium loading capacity of mitochondria, as well as mitochondrial morphology. Altogether, these findings suggest that PrxV is a multifunctional protein, which is essential for protection against apoptosis induced by anticancer drugs.


Subject(s)
Apoptosis , Carcinoma/enzymology , Lung Neoplasms/enzymology , Peroxidases/metabolism , Apoptosis/drug effects , Apoptosis/radiation effects , Calcium/metabolism , Carcinoma/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Down-Regulation , Humans , Lung Neoplasms/metabolism , Membrane Potentials , Mitochondria/metabolism , Mitochondria/ultrastructure , Peroxidases/physiology , Peroxiredoxins
19.
Inhal Toxicol ; 18(1): 79-92, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16326404

ABSTRACT

Inhaled cigarette smoke induces oxidative stress in the epithelium of airways. Peroxiredoxin V (PRXV) is a potent antioxidant protein, highly expressed in cells of the airway epithelium. The goal of our study was to determine whether cigarette smoke extract (CSE) influenced expression of this protein in airway epithelia in vivo and in vitro. In Sprague-Dawley rats, we determined effects of CSE on airway epithelial permeability, mRNA levels and expression of PRXV protein. Exposure of isolated tracheal segment in vitro to 20% CSE for 4 h resulted in development of increased permeability to albumin, significantly reduced mRNA levels for PRXV, and reduced amounts of PRXV protein in the epithelium. In cultures of the airway epithelial cell lines (Calu-3, JME), primary airway cell culture (cow), and alveolar epithelial cells A549, CSE also significantly decreased transepithelial electrical resistance and expression of PRXV protein, and induced glutathione and protein oxidation. To demonstrate functional importance of PRXV, we exposed clones of HeLa cells with siRNA-downregulated PRXV to hydrogen peroxide, which resulted in increased rate of cell death and protein oxidation. CSE directly downregulates expression of functionally important antioxidant enzyme PRXV in the epithelial cells of airways, which represents one pathophysiological mechanism of cigarette smoke toxicity.


Subject(s)
Nicotiana/adverse effects , Peroxidases/genetics , Smoke/adverse effects , Trachea/drug effects , Animals , Apoptosis , Electric Impedance , Epithelial Cells/metabolism , Gene Expression Regulation , Male , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Oxidative Stress , Permeability , Peroxiredoxins , Proteins/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Trachea/metabolism
20.
Biochem Biophys Res Commun ; 336(3): 762-9, 2005 Oct 28.
Article in English | MEDLINE | ID: mdl-16153605

ABSTRACT

During G0 phase the p130, member of the pRb tumor suppressor protein family, forms a repressor complex with E2F4 which is inactivated in G1/S by hyperphosphorylation of the p130. The role of p130 after G1/S remains poorly investigated. We found that in nuclear extracts of T98G cells, the p130-E2F4-DNA (pp-E2F4) complex does not dissociate at G1/S transition, but instead reverts to the p130-E2F4-cyclin E/A-cdk2 (cyc/cdk-pp-E2F4) complex, which is detected in S and G2/M phases of the cell cycle. Hyperphosphorylation of the p130 at G1/S transition is associated with a decrease of its total amount; however, this protein is still detected during the rest of the cell cycle, and it is increasingly hyperphosphorylated in the cytosol, but continuously dephosphorylated in the nucleus. Both nuclear and cytosol cell fractions in T98G cells contain a hyperphosphorylated form of p130 in complex with E2F4 at S and G2/M cell cycle phases. In contrast to T98G cells, transformation of the p130 containing cyc/cdk-pp-E2F4 complex into the p130-pp-E2F4 repressor does not occur in HeLa cells under growth restriction conditions.


Subject(s)
Cell Cycle , DNA-Binding Proteins/metabolism , Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , CDC2-CDC28 Kinases/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclin-Dependent Kinase 2 , Cytoplasm/metabolism , E2F4 Transcription Factor , HeLa Cells , Humans , Kinetics , Phosphorylation , Retinoblastoma-Like Protein p130
SELECTION OF CITATIONS
SEARCH DETAIL
...