Skip to main content
Erschienen in: Respiratory Research 1/2010

Open Access 01.12.2010 | Research

Ovalbumin sensitization and challenge increases the number of lung cells possessing a mesenchymal stromal cell phenotype

verfasst von: J Kelley Bentley, Antonia P Popova, Paul D Bozyk, Marisa J Linn, Amy E Baek, Jing Lei, Adam M Goldsmith, Marc B Hershenson

Erschienen in: Respiratory Research | Ausgabe 1/2010

Abstract

Background

Recent studies have indicated the presence of multipotent mesenchymal stromal cells (MSCs) in human lung diseases. Excess airway smooth muscle, myofibroblasts and activated fibroblasts have each been noted in asthma, suggesting that mesenchymal progenitor cells play a role in asthma pathogenesis. We therefore sought to determine whether MSCs are present in the lungs of ovalbumin (OVA)-sensitized and challenged mice, a model of allergic airways disease.

Methods

Balb/c mice were sensitized and challenged with PBS or OVA over a 25 day period. Flow cytometry as well as colony forming and differentiation potential were used to analyze the emergence of MSCs along with gene expression studies using immunochemical analyses, quantitative polymerase chain reaction (qPCR), and gene expression beadchips.

Results

A CD45-negative subset of cells expressed Stro-1, Sca-1, CD73 and CD105. Selection for these markers and negative selection against CD45 yielded a population of cells capable of adipogenic, osteogenic and chondrogenic differentiation. Lungs from OVA-treated mice demonstrated a greater average colony forming unit-fibroblast (CFU-F) than control mice. Sorted cells differed from unsorted lung adherent cells, exhibiting a pattern of gene expression nearly identical to bone marrow-derived sorted cells. Finally, cells isolated from the bronchoalveolar lavage of a human asthma patient showed identical patterns of cell surface markers and differentiation potential.

Conclusions

In summary, allergen sensitization and challenge is accompanied by an increase of MSCs resident in the lungs that may regulate inflammatory and fibrotic responses.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1465-9921-11-127) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

JKB contributed aspects of all experimental designs, analyses and manuscript preparation; APP contributed to conceptual analyses of experiments; PDB contributed to conceptual analyses of gene chip experiments; MJL performed tissue processing, sectioning, and immunostaining; AEB assisted in developing differentiation assays; JL performed Western analyses; AMG performed ELISA and multiplex analyses; MBH contributed to conceptual design and analyses of experiments, manuscript preparation, and funding. All authors read and approved the final manuscript.

Introduction

Adult bone marrow contains a minority population of mesenchymal stem cells that are thought to contribute to the regeneration of connective tissues such as bone, cartilage, muscle, ligaments, tendons, fat and stroma [1]. These cells demonstrate cell surface markers consistent with a mesenchymal origin (e.g., Stro-1, Sca-1, CD73, CD90, CD105) but fail to express markers associated with a hematopoietic (CD34, CD45) or endothelial cell (CD31) origin. Subsequently, multipotent mesenchymal stem cells were isolated from peripheral sites, including peripheral blood [2], adipose tissue [3], articular synovium [4] and trabecular bone [5]. More recently, it has been suggested that most organs apparently carry their own population of pluripotent mesenchymal stem cells in a perivascular compartment that participate in tissue repair [6]. The perivascular niche has been demonstrated to be the source of mesenchymal stem cells in adipose tissue [7], skeletal muscle [8] and other organs [9].
With regard to the lung, mesenchymal stem cells have been isolated from the bronchial tissue of patients undergoing lobectomy for primary lung tumors [10]. Our laboratory has isolated mesenchymal stem cells from the tracheal aspirates of premature infants undergoing mechanical ventilation for respiratory distress [11]. Mesenchymal stem cells of donor sex identity have been found in lung allografts years after transplantation, suggesting that these stromal cells may originate from the lung tissue itself, perhaps from the perivascular compartment [12]. Finally, we [13] and others [14, 15] have shown that, in addition to the above cell types, mesenchymal stem cells may undergo differentiation to myofibroblasts.
Recent work suggests that there is a hierarchy of multipotent mesenchymal stromal cells ranging from true self-renewing stem cells with multilineage differentiation capacity to those with more restricted differentiation potential, until a state of complete restriction to the fibroblast is reached [16]. Since the clonogenicity or self-renewal of most mesenchymal stem cell isolates has not been thoroughly tested, we now refer to cells with the surface markers and differentiation potential of mesenchymal stem cells as mesenchymal stromal cells (MSCs), which have a more restricted differentiation potential.
There is abundant evidence in the literature suggesting that mesenchymal progenitor cells are involved in asthma pathogenesis. Excess airway smooth muscle, caused in part by an increase in the number of smooth muscle cells, has been well-described in asthma [17, 18]. Patients with severe asthma also demonstrate an accumulation of myofibroblasts in the airway subepithelium [1922]. These alterations may extend beyond the central airways to the distal airways and lung parenchyma [23]. Finally, in contrast to control subjects, "activated" fibroblasts have been identified in the bronchoalveolar lavage (BAL) fluid of patients with asthma [24]. We therefore hypothesized that multipotent MSCs, which may serve as progenitor cells of smooth muscle cells and myofibroblasts, are increased in the lungs of ovalbumin-sensitized and -challenged mice, an animal model of allergic airways disease. Previous studies have demonstrated hyperplasia of myofibroblasts in both the large and small airways of these mice, including the terminal bronchioles, alveolar ducts and alveolar walls [25].

Materials and methods

OVA-sensitization and challenge

Mice (BALB/cByJ, Jackson Labs, Bar Harbor, ME) were sensitized to sterile LPS-free OVA (Pierce, Rockford, IL) or PBS control by intraperitoneal injection and serially challenged over a month with intranasal instillations of OVA or PBS (Figure 1), as previously described [26, 27]. This study was approved by the Institutional Animal Care and Use Committee.

Flow cytometry of whole lung digests and adherent cells

Lungs from PBS- and OVA-treated mice were perfused with 5 mM EDTA, minced into 1-2 mm blocks and digested for 1 hr in 5 mg/ml collagenase. Cells were obtained by filtration through a 70 μm mesh. Red blood cells were lysed with an ammonium chloride buffer. Cells were washed in 10% serum-containing DMEM with 4 mM glucose, 1 mM sodium pyruvate, and 2 mM L-glutamine and plated at 106 cells/100 mm dish. After 5 days, adherent cells were removed from the plates by trypsin, washed with Ca2+, Mg2+-containing PBS and blocked with 1% BSA in PBS for 20 min at 4°C. Within three hours after immunostaining (below), cells were sorted using a BD Biosciences FACSDiVa High-Speed Cell Sorter (San Diego, CA) with 350 nm, 488 nm, and 633 nm lasers. Cells were stained with 2.5 μg/mL of FITC- or AlexaFluor (AF)-conjugated antibodies. Typically, anti-Stro-1 was conjugated to AF 350, anti-CD34 was conjugated to FITC, anti-CD105 was coupled to AF555, anti-CD73 was coupled to AF633 and anti-CD45 was conjugated to AF750. Cells were negatively selected for CD34 and CD45 and positively selected for Stro-1, CD73 and CD105 binding (see text). Cells were collected in 1 mL DMEM supplemented with 10% FBS and cultured for 2 weeks to allow colony formation before expansion for study.

Determination of colony forming unit-fibroblast (CFU-F)

Lungs from PBS- and OVA-treated mice were perfused with 5 mM EDTA, minced into 1-2 mm blocks and digested for 1 hr in 5 mg/ml collagenase. Cells were obtained by filtration through a 70 μm mesh. Trypan blue-excluding cells from each animal were serially diluted and plated in DMEM with 10% FBS at 100 cells/well in 2 mL of medium into a 30 mm well of a 6 well dish (in triplicate). Cells were allowed to grow in 10% FBS/DMEM for 14 days. After 14 days, plates were stained with 0.5% toluidine blue for 5 min, washed three times with PBS, and independent colonies of primary mouse lung fibroblasts were counted, as described previously [28]. A similar procedure was followed to determine the CFU-F of sorted Stro-, CD73, CD105-positive mouse lung MSCs.

Analysis of cultured sorted cells from the lungs of OVA-sensitized and challenged mice

Cells (passage 3-5 post-sort) were dissociated from plastic using cell dissociation buffer (PBS supplemented with 5 mM EDTA) and analyzed for the surface markers described above, as well as CD90.2 (rat anti-mouse Thy 1.2, clone 30-H12, BD Pharmingen) and Sca-1 (rat anti-mouse Ly 6-A/E, clone e13-161.7, BD Pharmingen). Cells were typically stained with 2 μg/mL FITC- or AF488-conjugated antibodies or isotypic control immunoglobulins and 50 μg/mL propidium iodide.

Fluorescence microscopy

The pulmonary artery was perfused with EDTA. Mouse lung vasculature was perfused with 5 mM EDTA in PBS injected through the pulmonary artery until the lung blanched. Lungs were inflated to 30 cm H2O pressure with 4% paraformaldehyde (Sigma-Aldrich, St. Louis, MO) and placed in formalin overnight. Paraffin-embedded lungs were sectioned with 5 μm-thick sections. AlexaFluor (AF) dye antibody conjugates were prepared using N-hydroxy succinimide esterified dye conjugation reactions with 20 μg antibody and 100 μg activated dye in 10 mM sodium bicarbonate buffer (pH 8.5) for 1 hour. The reaction was quenched by making it 500 mM Tris HCl pH 7.2 for 1 hour, and the conjugated antibody was purified by separating it from quenched dye over a G-50 spin column (GE Healthcare, Piscataway, NJ). Slides were probed with 2 μg/mL AF or fluorescein isothiocyanate (FITC)-conjugated anti-Stro-1 (clone STRO-1, R&D Systems, Minneapolis, MN), anti-CD34 clone MEC14.7, anti-CD45 clone 30-F11, anti-CD73 clone TY/11.8, anti CD105 clone MJ7/18 (each from BioLegend, San Diego, CA) and Cy3- or FITC-conjugated mouse anti-α-smooth muscle actin (clone 1A4, Sigma-Aldrich). Nuclei were visualized with 10 μg/mL Hoechst 33258 (Sigma-Aldrich). Cells were imaged using either an Olympus XI74 microscope (Center Valley, PA) for epifluorescence, a Zeiss Axioplan microscope equipped with an ApoTome and digital AxioCamMR CCD camera, or a Zeiss LSM 150 confocal microscope (Thornwood, NY).

Differentiation of clonal MSCs

Passage two sorted cells were serially diluted in 96 well plates to 0.3 cells per well. After 2 weeks in this media, individual colonies were trypsinized, dispersed into 100 mm dishes, allowed to grow to confluence and plated on fibronectin-coated slides. Myogenic differentiation was induced by addition of 10 ng/mL transforming growth factor (TGF)-β, 1 μg/mL insulin, 0.55 μg/mL transferrin and 670 ng/mL selenium (Invitrogen, Carlsbad, CA) for 6 days. These cells were immunostained for α-actin (clone 1A4, Sigma-Aldrich) and myosin heavy chain (clone hSM-V, Sigma-Aldrich). Adipogenic differentiation was induced by culture in DMEM supplemented with 5% fetal bovine serum (FBS), 10 μM dexamethasone, 50 μM indomethacin, 100 μM isobutylmethylxanthine (IBMX) and 10 μg/mL insulin for 21 days, as described [11] Formalin-fixed cells were stained for neutral lipids with Oil Red O. Osteogenic differentiation was accomplished by culture in DMEM supplemented with 5% FBS, 100 nM dexamethasone, 10 mM β-glycerophosphate and 50 μg/mL ascorbic acid for 21 days, as described [11]. Formalin-fixed cells were stained for calcium phosphate with alizarin red.
To further assess differentiation state, cells were solubilized and RNA extracted using a guanidine isothiocyanate-phenol protocol (Trizol, InVitrogen). Quantitative two-step real time PCR for differentiation marker transcripts was performed using an Eppendorf realplex2 (Westbury, NY). Transcripts were examined for myocardin (MYOCD, forward sequence, CCCATGGACTCTGCCTATGC; reverse sequence, CTGAGGATCTGTAGCTGCAGGAAT), smooth muscle myosin heavy chain (MHC11, forward sequence, GCCGACACAGCCTACAGAAG; reverse sequence, TTTCCGGCTCCAGACTCA), transgelin (SM22, forward sequence, GCCAACAAGGGTCCATCCTA; reverse sequence, CTCCAGCTCCTCGTCATACTTC), peroxisome proliferator-activated receptor (PPAR)-γ (forward sequence, GGCCCACCAACTTCGGAATC; reverse sequence, GTAAAGGGCTTGATGTCAAAGGAA), fatty acid binding protein 4/adipocyte protein-2 (FABP4, forward sequence, GGCTTTGCCACAAGGAAAGTG; reverse sequence, CGCCCAGTTTGAAGGAAATCTC), osteopontin/secreted phosphoprotein 1 (SSP1, forward sequence, CTCACCATTCGGATGAGTCTGAT; reverse sequence, AGGGACGATTGGAGTGAAAGTG) and osteocalcin/bone γ-carboxyglutamate (gla) protein (BGLAP, forward sequence, CAGCTTGGCCCAGACCTA; reverse sequence, GCCAGGGTCAGCAGAGTG), Expression levels were normalized to that of glyceraldehyde 3-phosphate dehydrogenase (GAPDH; forward sequence, TCCACTCACGGCAAATTCAAC); reverse sequence, CGCTCCTGGAAGATGGTGATG).

Western analyses

Mouse lung MSCs were washed in PBS, homogenized in 50 mM Tris (pH 7.5), 100 mM NaCl, 50 mM NaF, 40 mM β-glycerophosphate, 2 mM EDTA, 200 μM Na3VO4, and 1% Triton X-100 containing complete protease inhibitors (Roche Diagnostics, Indianapolis, IN). After centrifugation at 10,000 g for 30 min to remove nuclei and cell particles, supernatants were resolved by SDS-PAGE and transferred to nitrocellulose. Membranes were probed with mouse monoclonal anti-α-smooth muscle actin (MAb 1A4; Calbiochem, San Diego, CA), mouse monoclonal anti-myosin heavy chain (MHC) (MAb HSM-V; Sigma-Aldrich, St. Louis, MO) or mouse monoclonal anti-β-actin (Sigma). Antibody binding was detected in Westerns with a peroxidase-conjugated anti-rabbit or anti-mouse IgG and chemiluminescence (Thermo-Pierce, Rockford, IL).

Gene arrays

We examined the gene expression profile of MSCs using the Illumina MouseWG-6 expression BeadChip platform (San Diego, CA). This system detects 26,766 coding transcripts with well-established annotations, 6,277 coding transcripts with provisional annotations and 56 non-coding transcripts with well-established annotations. Total RNA was extracted using the RNeasy Plus Mini kit (Qiagen, Valencia, CA). Further preparation and analysis was carried out by the University of Michigan Sequencing Core, according to the chip manufacturer's recommended protocol. Hybridized biotinylated cRNA was detected with streptavidin-Cy3 and quantitated using an Illumina BeadArray Reader. The gene expression profile of lung MSCs was compared to primary mouse lung fibroblasts as well as primary mouse bone marrow MSCs, which were isolated from the femurs of OVA-treated mice by flow cytometry, using gates identical to those used for lung MSCs.
For microarray statistical analysis, background-corrected values for each probe on the BeadChip array were extracted using GenomeStudio Data Analysis Software (Illumina). Detection p-values were computed using a non-parametric method. Probe signals are ranked relative to signals of negative controls. Detection p-value = 1-R/N, where R is the rank of the gene signal relative to negative controls and N is number of negative controls. Statistical differences in gene expression between lung and bone marrow-derived cells were calculated using the Illumina Custom differential expression algorithm. Multiple tests were corrected by the Benjamini and Hochberg false discovery rate [29]. For qPCR, data were normalized for GAPDH, and differences in gene expression analyzed by paired t test.

Isolation and characterization of MSCs from the BAL of a patient with asthma

This study was approved by the University of Michigan Institutional Review Board. After informed consent, BAL fluid was obtained from three pediatric asthma patients who were undergoing diagnostic flexible bronchoscopy. Cells were sedimented for 5 min at 500 × g, washed in media, counted, and serial dilutions of 100,000 to 100 cells were plated and allowed to grow for 21 days CellCe, Single colonies were isolated, expanded, and assessed for cell surface markers and pluripotency. Cell surface markers and differentiation potential were compared to human neonatal lung MSCs [11] and primary human lung fibroblasts (Lonza, Walkersville, MD). Cell surface markers were examined by flow cytometry using human-specific antibodies (BioLegend).. Cells were differentiated along adipogenic and osteogenic lines as described above. For chondrogenic differentiation, cells were differentiated along cartilage lines by first sedimenting (10 min at 500 × g) 200,000 trypsinized cells in a sterile 15 mL polypropylene culture tube and maintaining them as a pellet for 21 days in serum-free DMEM composed with 10 ng TGF-β1 and 10 ng bone morphogenetic protein-4/mL [30]. At the end of this time cell pellets were fixed overnight in formalin, processed for paraffin embedding and sectioning, and stained for chondroitin sulfate with Alcian blue[31]. A nuclear fast red counterstain was used to help distinguish nuclei acids from acid mucopolysaccharides.

Results

OVA sensitization and challenge is associated with the appearance of Stro-1, CD73, and CD105-positive cells with high colony forming potential

We used flow cytometry to examine cells for cell surface markers typically associated with MSCs (Figure 2). Cells from whole lung digests were plated on plastic for five days to enrich for adherent lung fibroblast cells. We stained live cells from PBS- and OVA-treated mice for Stro-1, CD73, CD105, CD34 and CD45. We found a subpopulation of fibroblast-like cells from OVA-treated mice which showed greater Stro-1 expression (Figure 2A). We found distinct side populations of cells from OVA-treated mice which showed greater CD105 expression (Figures 2B and 2C), Both populations gated in the same forward scattering area. To eliminate hematopoietic cells, CD34 and CD45-positive cells were delineated by the windows shown (Figures 2B and 2C) and further gated for CD73 and CD105 expression, leaving a population of Stro-1-, CD73- and CD105-positive cells (Figure 2D). This population represents about 0.04% of the original lung cell digest (Table 1) and is here designated a lung MSC population. These cells were rarely detectable in PBS-treated lungs (0.004%).
Table 1
Populations of Stro-1-, CD105-positive, CD34-CD45-negative cells from PBS- and OVA-treated lungs.
Population of Cells Analysed
PBS
OVA
Total population
100,000
100,000
High Stro-1, low CD34, low CD45, low CD73, high CD105
14 ± 7
36 ± 19
High Stro-1, low CD34, low CD45, medium CD73, high CD105
12 ± 6
138 ± 61
High Stro-1, low CD34, low CD45, high CD73, high CD105
4 ± 1
41 ± 13*
Cells obtained and sorted as described above were counted from 3 sets of 5 PBS- or OVA-treated mice (*P < 0.05, unpaired t-test).
The potential of adherent cells to form colonies was assessed by colony-forming unit-fibroblast (CFU-F) assay [28]. Sorted cells and primary mouse lung fibroblasts were serially diluted and 100 cells were plated in DMEM with 10% FBS. After 14 days, plates were fixed and stained. Sorted cells demonstrated a CFU-F of 29.0 ± 1.4 (mean ± SEM, n = 4) out of 100 cells plated, significantly greater than unsorted lung cells from either PBS (1.5 ± 0.1)- or OVA-treated (8.9 ± 2.2) mice (n = 6 for both groups, P < 0.05, ANOVA).
Sorted and clonal cells were trypsinized after two further passages and processed for flow cytometry using the same antibodies with which they were initially sorted. The sorted cells retained MSC cell surface antigens after propagation, and also stained positively for CD90.2 and Sca-1 (Figure 3). In the mouse lung, the CD45-, CD31-negative, Sca-1-positive phenotype has been associated with the mesenchymal progenitor cell lineage [32]
We examined sections from PBS- and OVA-treated mouse lungs for surface markers typically associated with MSCs including Stro-1, CD73 and CD105. In humans, Stro-1 is almost exclusively found on bone marrow-derived stromal cells [33]. Stro-1 localization has also been noted in mouse mesenchymal cells of the bone marrow [34] and developing periodontium [35]. CD73 (SH3/SH4) and CD105 (SH2/endoglin) are localized at the plasma membrane of bone marrow mesenchymal stem cells [36]. A small number of peribronchial cells stained positively for Stro-1, CD73, CD105 and α-smooth muscle actin (Figure 4).

Sorted Stro-1(+), CD73(+), CD105(+) cells may be differentiated along a variety of mesenchymal lineages

Cells from Stro-1-, CD73-, CD105-positive, CD34-, CD45-negative subpopulations were seeded onto plastic dishes. Only the high CD73, high CD105 cells grew well and were studied further. To isolate individual clones, passage two sorted cells were serially diluted in 96 well plates to 0.3 cells per well. After two weeks of growth in this media, wells with cells were trypsinized. Cells were dispersed into 100 mm dishes, allowed to grow to confluence, plated on fibronectin-coated slides and differentiated along adipogenic, osteogenic and myofibroblast lines (Figures 5, 6). When cells are treated with adipogenic medium (10 μM dexamethasone, 100 μM IBMX, 50 μM indomethacin and 100 μg/mL insulin), cells accumulated neutral lipid vesicles (Figure 5A) and expressed more mRNA encoding PPARG and FABP4 (Figure 5B). Osteogenic differentiation with 0.1 μM dexamethasone, 10 mM β-glycerophosphate and 50 μg/ml ascorbic acid caused cells to increase calcium phosphate deposition as well as mRNA expression of SSP1 and BGLAP. TGF-β treatment induced expression of MYOCD, MHC11, and TAGLN (Figure 6A). Western analyses of these cells demonstrated significantly more α-actin and MHC (Figure 6B). The size of α-actin-positive cells was increased, as assessed by increased forward scatter (Figure 6C). When cells were treated with TGF-β, many differentiated into filamentous α-actin- and MHC-expressing myofibroblasts (Figure 6D). On the basis of their cell surface markers, ability form colonies, and differentiation potential, these cells meet established criteria for MSCs [37].

Sorted lung MSCs have a significantly different gene expression pattern from either unsorted lung fibroblasts

To better characterize the differences between sorted lung MSCs and other resident fibroblast-like cells, mRNA samples from three different sorted clonal lines were used to analyse patterns of gene expression. Out of 33043 mouse mRNA transcripts, a detection p ≤ 0.05 for 12,331 genes of mouse lung MSCs was obtained. Global transcript expression concordance (r2) between the sorted lung MSCs and adherent unsorted lung fibroblasts was 0.7539 (Figure 7A). We found a differential p value of ≤0.05 for 5559 genes between sorted mouse lung MSCs and unsorted adherent lung fibroblasts. On the other hand, there was high concordance between lung and bone marrow MSCs (r2 = 0.9872) (Figure 7B). Thus, the sorted multipotent stromal cells represent a distinct subpopulation of cells within the lung.
Of the 5559 genes differing between lung MSCs and lung fibroblasts, 2480 were upregulated. These genes included selected cytokines and paracrine factors; receptors, signaling intermediates and transcriptional regulators of the TGF-β1 and TNF-α pathways; and receptor tyrosine kinases and their downstream signaling intermediates (Table 2 and Additional File 1, Table S1, supplement).
Table 2
Selected genes upregulated in sorted lung MSCs relative to lung fibroblasts.
Symbol average
Lung MSCs average
Lung fibroblast
Fold difference
Differential p-value
Secreted cytokines and paracrine factors
    
Prl2c3
20800
2.91
7147.77
1.00E-13
Prl2c4
16700
2.49
6706.83
1.01E-13
Prl2c2
16400
2.37
6919.83
1.01E-13
Efnb2
250
42.4
5.90
2.98E-08
Prl2c5
224
71.5
3.13
7.15E-08
Grem1
17700
2520
7.02
1.42E-06
Ptges3
2080
763
2.73
1.20E-05
Fgf10
3710
853
4.35
0.000233
Ctf1
265
109
2.43
0.000226
Ccl25
171
58.9
2.90
0.000239
Fgf7
7600
3580
2.12
0.00038
Efna4
176
85.3
2.06
0.00268
Bmper
2060
1020
2.02
0.0041
Lefty1
411
244
1.68
0.0163
Gdf11
52.3
7.81
6.70
0.0279
Thbs1
961
300
3.20
0.0293
Cx3cl1
384
81.6
4.71
0.0311
Sema3a
638
14.8
43.11
0.0367
Spp1
12500
8920
1.40
0.0469
TGF-β1 signaling
    
Smad2
334
86.4
3.87
5.67E-07
Smad3
889
410
2.17
5.67E-07
Mapk13
387
104
3.72
1.28E-06
Hdac2
2440
1300
1.88
1.58E-05
Trrap
207
54.2
3.82
4.02E-05
Hdac4
179
69.6
2.57
0.000433
Mapk11
115
30.3
3.80
0.000661
Tgfbrap1
603
195
3.09
0.00172
Nfatc3
287
121
2.37
0.00226
Hdac3
1080
377
2.86
0.00241
Hdac10
60.3
19.4
3.11
0.00378
Acvr2b
177
107
1.65
0.0308
Endogl1
77.5
39.1
1.98
0.0462
Abbreviations are: Prl2c3, encoding prolactin family 2, subfamily c, member 3; Prl2c4, prolactin family 2, subfamily c, member 4 ; Prl2c2. prolactin family 2, subfamily c, member 2; Efnb2, ephrin B2; Prl2c5, prolactin family 2, subfamily c, member 5; Grem1, gremlin 1; Ptges3, prostaglandin E synthase 3; Fgf10, fibroblast growth factor 10; Ctf1, cardiotrophin 1; Ccl25, chemokine (C-C motif) ligand 25; Fgf7, fibroblast growth factor 7 or keratinocyte growth factor; Efna4, ephrin A4; Bmper, BMP-binding endothelial regulator; Lefty1, left right determination factor 1; Gdf11, growth differentiation factor 11 (BMP 11); Thbs1, thrombospondin 1; Cx3cl1, chemokine (C-X3-C motif) ligand 1; Sema3a, sema domain, secreted, (semaphorin) 3A; Spp1, secreted phosphoprotein 1, osteopontin; Smad2, MAD homolog 2 (Drosophila); Smad3, MAD homolog 3 (Drosophila); Mapk13, mitogen activated protein kinase 13 (p38); Hdac2, histone deacetylase 2; Trrap, transformation/transcription domain-associated protein; Hdac4, histone deacetylase 4; Mapk11, mitogen-activated protein kinase 11 (p38 δ); Tgfbrap1, transforming growth factor, beta receptor associated protein 1; Nfatc3, nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 3; Hdac3, histone deacetylase 3; Hdac10, histone deacetylase 10; Acvr2b, activin receptor IIB; Endogl1, endoglin (CD105);

Isolation of mesenchymal progenitor cells with surface markers of MSCs from a patient with asthma

We have isolated pluripotent mesenchymal cells from the tracheal aspirates of premature infants undergoing mechanical ventilation for respiratory distress [11]. Activated fibroblasts have previously been isolated from human asthma patients [24]. We examined the BAL fluid of three children with asthma who were undergoing diagnostic flexible bronchoscopy. After two weeks of culture, one sample yielded colonies of adherent cells. These cells stained low for CD31, CD34 or CD45 and high for Stro-1, CD73, CD105, and CD166 (Figure 8A). Unlike normal human lung fibroblasts, but similar to pluripotent mesenchymal cells from human neonates [11], these cells could be differentiated along adipogenic, osteogenic and chondrogenic lines (Figure 8B). Together, these data suggest that pluripotent mesenchymal cells may be identified in the BAL of human asthma patients.

Discussion

Mesenchymal stem cells are multipotent cells capable of differentiation into a select range of mesenchymal cell types influenced by their microenvironment. They are identified by their cell surface markers, high proliferation rate, ability to form colonies and differentiation potential [37]. More recent work shows that mesenchymal stem cells are not after all uniform, and that pluripotent mesenchymal cells may be separated into a hierarchy with differential self-renewal potential and multipotency [16].
Using cell sorting, we isolated a population of CD34- and CD45-negative, Stro-1-positive, high CD73- and CD105-positive cells. In humans, Stro-1 is almost exclusively found on bone marrow-derived MSCs [33]. Stro-1 has been described in bone marrow-derived stem cells in the mouse [34]. CD73 (SH3/SH4) and CD105 (SH2/endoglin) are localized at the plasma membrane of bone marrow mesenchymal stromal cells [36]. The CD34- and CD45-negative, Stro-1-positive, high CD73- and CD105-positive cells grew on plastic, demonstrated ample colony-forming potential, and were capable of differentiation to osteogenic, adipogenic, and myogenic phenotypes. Taken together, these data suggest that OVA-sensitization and -challenge increases the population of MSCs in the lung.
While we did not determine the precise function of MSCs in allergic airways disease, we consider two possibilities to be most likely. First, MSCs could constitute a source of airway myofibroblasts, leading to increased airway smooth muscle mass. Patients with severe asthma also demonstrate an accumulation of myofibroblasts in the airway subepithelium [1921]. These alterations may extend beyond the central airways to the distal airways and lung parenchyma [23]. After allergen challenge, myofibroblasts in the subepithelium of the asthmatic airway may migrate to the smooth muscle layer [22]. Previous studies have noted increased airway smooth muscle cell number in fatal and non-fatal asthma [17, 18], as well as OVA-sensitized and -challenged mice [38]. Mesenchymal stem cells generally express low levels of α-smooth muscle actin, but may express high levels of this contractile protein upon TGF-β treatment [11]. In multipotent stem cells from tracheal aspirates of premature infants, autocrine production of TGF-β further drives myofibroblastic differentiation,[13]. Finally, sorted lung MSCs were enriched for many transcripts which could play a role in myofibroblast differentiation and fibrosis including those encoding CD105/endoglin, part of the TGF-β receptor complex; the TGF-β family protein Gdf11 [39, 40]; the TGF-β antagonists Gremlin, Lefty1 and BMP-binding endothelial regulator [41, 42]; SMADs 2 and 3; NFATc3, a transcription factor involved in TGF-β1 signaling in airway smooth muscle [43]; and osteopontin which has been shown to be required for TGF-β expression and lung fibrosis in bleomycin- [44] and OVA-treated mice [45].
Another possible function of resident multipotent stromal cells is immunomodulation. Lung-resident mesenchymal stem cells from human lung allografts inhibit T cell proliferation in vitro [46]. Intravenously-administered murine mesenchymal stem cells derived from plastic-adherent bone marrow cells protect against bleomycin lung injury through the expression of interleukin-1 receptor antagonist [4749]. In addition, intratracheal pluripotent mesenchymal stem cells have been shown to decrease endotoxin-induced lung injury [50]. In the present study, sorted lung MSCs were enriched in transcripts encoding the pro-inflammatory IL-6 family member cardiotrophin. This cytokine also is involved in ASM hypertrophy [43]. In addition, cultured lung multipotent stromal cells also produced MCP-1/CCL2 and KC/CXCL1, cytokines that would be expected to increase airway inflammation. Several members of the prolactin 2c gene family were greatly enriched in lung multipotent stromal cells. While humans possess only one prolactin gene used for both endocrine and paracrine functions, the prolactins are a multigene family in mice [51, 52]. The prolactin-like peptides, also referred to as proliferins, have been implicated in wound-healing [53], angiogenesis [54] and B cell tolerance [55].
We successfully isolated MSCs from a patient with asthma, proof of concept that MSCs may be isolated from the BAL of these patients. As noted above, multipotent mesenchymal stem cells have been isolated from the tracheal aspirates of premature infants undergoing mechanical ventilation for respiratory distress [11] and the bronchoalveolar lavage of patients who have undergone lung transplantation [12]. In the latter studies, cells were grown from colonies identified 7-21 days after initial plating on plastic [11, 12]. In the present study, MSCs were identified by flow cytometry after only 5 days of adherence to plastic, and by fluorescence microscopy at the time of allergen challenge. Together, these data provide stronger evidence that multipotent mesenchymal cells are present in the lung in vivo. However, at this time it is premature to say whether MSCs play a pathogenic role in human asthma.
In summary, we have found that allergen-sensitization and -challenge leads to the emergence of a lung cell population with the surface markers, differentiation potential and clonogenic potential of multipotent stromal cells. Further animal studies will be required to determine the physiologic role of these cells. Finally, additional work is needed to determine whether lung MSCs play a role in human asthma.

Acknowledgements

This work was supported by National Institutes of Health grants HL079339 and HL090134 (M.B.H.)
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

JKB contributed aspects of all experimental designs, analyses and manuscript preparation; APP contributed to conceptual analyses of experiments; PDB contributed to conceptual analyses of gene chip experiments; MJL performed tissue processing, sectioning, and immunostaining; AEB assisted in developing differentiation assays; JL performed Western analyses; AMG performed ELISA and multiplex analyses; MBH contributed to conceptual design and analyses of experiments, manuscript preparation, and funding. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak D: Multilineage Potential of Adult Human Mesenchymal Stem Cells. Science 1999, 284:143–147.CrossRefPubMed Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak D: Multilineage Potential of Adult Human Mesenchymal Stem Cells. Science 1999, 284:143–147.CrossRefPubMed
2.
Zurück zum Zitat Zvaifler NJ, Marinova-Mutafchieva L, Adams G, Edwards CJ, Moss J, Burger JA, Maini RN: Mesenchymal precursor cells in the blood of normal individuals. Arthritis Res 2000, 2:477–488.CrossRefPubMedPubMedCentral Zvaifler NJ, Marinova-Mutafchieva L, Adams G, Edwards CJ, Moss J, Burger JA, Maini RN: Mesenchymal precursor cells in the blood of normal individuals. Arthritis Res 2000, 2:477–488.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH: Human Adipose Tissue Is a Source of Multipotent Stem Cells. Mol Biol Cell 2002, 13:4279–4295.CrossRefPubMedPubMedCentral Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH: Human Adipose Tissue Is a Source of Multipotent Stem Cells. Mol Biol Cell 2002, 13:4279–4295.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat De Bari C, Dell'Accio F, Tylzanowski P, Luyten FP: Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis Rheum 2001, 44:1928–1942.CrossRefPubMed De Bari C, Dell'Accio F, Tylzanowski P, Luyten FP: Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis Rheum 2001, 44:1928–1942.CrossRefPubMed
5.
Zurück zum Zitat Tuli R, Tuli S, Nandi S, Wang ML, Alexander PG, Haleem-Smith H, Hozack WJ, Manner PA, Danielson KG, Tuan RS: Characterization of Multipotential Mesenchymal Progenitor Cells Derived from Human Trabecular Bone. Stem Cells 2003, 21:681–693.CrossRefPubMed Tuli R, Tuli S, Nandi S, Wang ML, Alexander PG, Haleem-Smith H, Hozack WJ, Manner PA, Danielson KG, Tuan RS: Characterization of Multipotential Mesenchymal Progenitor Cells Derived from Human Trabecular Bone. Stem Cells 2003, 21:681–693.CrossRefPubMed
6.
Zurück zum Zitat Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS, Andriolo G, Sun B, Zheng B, Zhang L, et al.: A Perivascular Origin for Mesenchymal Stem Cells in Multiple Human Organs. Cell Stem Cell 2008,3(3):301–313.CrossRefPubMed Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS, Andriolo G, Sun B, Zheng B, Zhang L, et al.: A Perivascular Origin for Mesenchymal Stem Cells in Multiple Human Organs. Cell Stem Cell 2008,3(3):301–313.CrossRefPubMed
7.
Zurück zum Zitat Zannettino AC, Paton S, Arthur A, Khor F, Itescu S, Gimble JM, Gronthos S: Multipotential human adipose-derived stromal stem cells exhibit a perivascular phenotype in vitro and in vivo. J Cell Physiol 2008, 214:413–421.CrossRefPubMed Zannettino AC, Paton S, Arthur A, Khor F, Itescu S, Gimble JM, Gronthos S: Multipotential human adipose-derived stromal stem cells exhibit a perivascular phenotype in vitro and in vivo. J Cell Physiol 2008, 214:413–421.CrossRefPubMed
8.
Zurück zum Zitat Tare RS, Babister JC, Kanczler J, Oreffo RO: Skeletal stem cells: Phenotype, biology and environmental niches informing tissue regeneration. Mol Cell Endocrinol 2008, 288:11–21.CrossRefPubMed Tare RS, Babister JC, Kanczler J, Oreffo RO: Skeletal stem cells: Phenotype, biology and environmental niches informing tissue regeneration. Mol Cell Endocrinol 2008, 288:11–21.CrossRefPubMed
9.
Zurück zum Zitat da Silva Meirelles L, Chagastelles PC, Nardi NB: Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci 2006, 119:2204–2213.CrossRefPubMed da Silva Meirelles L, Chagastelles PC, Nardi NB: Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci 2006, 119:2204–2213.CrossRefPubMed
10.
Zurück zum Zitat Sabatini F, Petecchia L, Tavian M, Jodon de Villeroche V, Rossi GA, Brouty-Boye D: Human bronchial fibroblasts exhibit a mesenchymal stem cell phenotype and multilineage differentiating potentialities. Lab Invest 2005, 85:962–971.CrossRefPubMed Sabatini F, Petecchia L, Tavian M, Jodon de Villeroche V, Rossi GA, Brouty-Boye D: Human bronchial fibroblasts exhibit a mesenchymal stem cell phenotype and multilineage differentiating potentialities. Lab Invest 2005, 85:962–971.CrossRefPubMed
11.
Zurück zum Zitat Hennrick KT, Keeton AG, Nanua S, Kijek TG, Goldsmith AM, Sajjan US, Bentley JK, Lama VN, Moore BB, Schumacher RE, et al.: Lung cells from neonates show a mesenchymal stem cell phenotype. Am J Respir Crit Care Med 2007, 175:1158–1164.CrossRefPubMed Hennrick KT, Keeton AG, Nanua S, Kijek TG, Goldsmith AM, Sajjan US, Bentley JK, Lama VN, Moore BB, Schumacher RE, et al.: Lung cells from neonates show a mesenchymal stem cell phenotype. Am J Respir Crit Care Med 2007, 175:1158–1164.CrossRefPubMed
12.
Zurück zum Zitat Lama VN, Smith L, Badri L, Flint AJ, Andrei AC, Murray S, Wang Z, Liao H, Toews GB, Krebsbach PH, et al.: Evidence for Tissue-Resident Mesenchymal Stem Cells in Human Adult Lung from Studies of Transplanted Allografts. J Clin Invest 2007,117(4):989–996.CrossRefPubMedPubMedCentral Lama VN, Smith L, Badri L, Flint AJ, Andrei AC, Murray S, Wang Z, Liao H, Toews GB, Krebsbach PH, et al.: Evidence for Tissue-Resident Mesenchymal Stem Cells in Human Adult Lung from Studies of Transplanted Allografts. J Clin Invest 2007,117(4):989–996.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Popova AP, Bozyk PD, Goldsmith AM, Linn MJ, Lei J, Bentley JK, Hershenson MB: Autocrine production of TGF-{beta}1 promotes myofibroblastic differentiation of neonatal lung mesenchymal stem cells. Am J Physiol Lung Cell Mol Physiol 2010, 298:L735–743.CrossRefPubMedPubMedCentral Popova AP, Bozyk PD, Goldsmith AM, Linn MJ, Lei J, Bentley JK, Hershenson MB: Autocrine production of TGF-{beta}1 promotes myofibroblastic differentiation of neonatal lung mesenchymal stem cells. Am J Physiol Lung Cell Mol Physiol 2010, 298:L735–743.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Russo FP, Alison MR, Bigger BW, Amofah E, Florou A, Amin F, Bou-Gharios G, Jeffery R, Iredale JP, Forbes SJ: The Bone Marrow Functionally Contributes to Liver Fibrosis. Gastroenterology 2006,130(6):1807–1821.CrossRefPubMed Russo FP, Alison MR, Bigger BW, Amofah E, Florou A, Amin F, Bou-Gharios G, Jeffery R, Iredale JP, Forbes SJ: The Bone Marrow Functionally Contributes to Liver Fibrosis. Gastroenterology 2006,130(6):1807–1821.CrossRefPubMed
15.
Zurück zum Zitat Jeon ES, Moon HJ, Lee MJ, Song HY, Kim YM, Cho M, Suh DS, Yoon MS, Chang CL, Jung JS, et al.: Cancer-Derived Lysophosphatidic Acid Stimulates Differentiation of Human Mesenchymal Stem Cells to Myofibroblast-Like Cells. Stem Cells 2008,26(3):789–797.CrossRefPubMed Jeon ES, Moon HJ, Lee MJ, Song HY, Kim YM, Cho M, Suh DS, Yoon MS, Chang CL, Jung JS, et al.: Cancer-Derived Lysophosphatidic Acid Stimulates Differentiation of Human Mesenchymal Stem Cells to Myofibroblast-Like Cells. Stem Cells 2008,26(3):789–797.CrossRefPubMed
16.
Zurück zum Zitat Sarugaser R, Hanoun L, Keating A, Stanford WL, Davies JE: Human Mesenchymal Stem Cells Self-Renew and Differentiate According to a Deterministic Hierarchy. PLoS One 2009,4(8):e6498.CrossRefPubMedPubMedCentral Sarugaser R, Hanoun L, Keating A, Stanford WL, Davies JE: Human Mesenchymal Stem Cells Self-Renew and Differentiate According to a Deterministic Hierarchy. PLoS One 2009,4(8):e6498.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Ebina M, Takahashi T, Chiba T, Motomiya M: Cellular hypertrophy and hyperplasia of airway smooth muscles underlying bronchial asthma. A 3-D morphometric study. Am Rev Respir Dis 1993,148(3):720–726.CrossRefPubMed Ebina M, Takahashi T, Chiba T, Motomiya M: Cellular hypertrophy and hyperplasia of airway smooth muscles underlying bronchial asthma. A 3-D morphometric study. Am Rev Respir Dis 1993,148(3):720–726.CrossRefPubMed
18.
Zurück zum Zitat Woodruff PG, Dolganov GM, Ferrando RE, Donnelly S, Hays SR, Solberg OD, Carter R, Wong HH, Cadbury PS, Fahy JV: Hyperplasia of Smooth Muscle in Mild to Moderate Asthma without Changes in Cell Size or Gene Expression. Am J Respir Crit Care Med 2004,169(9):1001–1006.CrossRefPubMed Woodruff PG, Dolganov GM, Ferrando RE, Donnelly S, Hays SR, Solberg OD, Carter R, Wong HH, Cadbury PS, Fahy JV: Hyperplasia of Smooth Muscle in Mild to Moderate Asthma without Changes in Cell Size or Gene Expression. Am J Respir Crit Care Med 2004,169(9):1001–1006.CrossRefPubMed
19.
Zurück zum Zitat Roche WR, Williams JH, Beasley R, Holgate ST: Subepithelial fibrosis in the bronchi of asthmatics. Lancet 1989, 1:520–524.CrossRefPubMed Roche WR, Williams JH, Beasley R, Holgate ST: Subepithelial fibrosis in the bronchi of asthmatics. Lancet 1989, 1:520–524.CrossRefPubMed
20.
Zurück zum Zitat Brewster CE, Howarth PH, Djukanovic R, Wilson J, Holgate ST, Roche WR: Myofibroblasts and subepithelial fibrosis in bronchial asthma. American Journal of Respiratory Cell & Molecular Biology 1990,3(5):507–511.CrossRef Brewster CE, Howarth PH, Djukanovic R, Wilson J, Holgate ST, Roche WR: Myofibroblasts and subepithelial fibrosis in bronchial asthma. American Journal of Respiratory Cell & Molecular Biology 1990,3(5):507–511.CrossRef
21.
Zurück zum Zitat Vignola AM, Chanez P, Chiappara G, Merendino A, Pace E, Rizzo A, laRocca AM, Bellia V, Bonsignore G, Bousquet J: Transforming growth factor-B expression in mucosa biopsies in asthma and chronic bronchitis. Am J Resp Crit Care Med 1997, 156:591–599.CrossRefPubMed Vignola AM, Chanez P, Chiappara G, Merendino A, Pace E, Rizzo A, laRocca AM, Bellia V, Bonsignore G, Bousquet J: Transforming growth factor-B expression in mucosa biopsies in asthma and chronic bronchitis. Am J Resp Crit Care Med 1997, 156:591–599.CrossRefPubMed
22.
Zurück zum Zitat Gizycki MJ, Adelroth E, Rogers AV, O'Byrne PM, Jeffery PK: Myofibroblast involvement in the allergen-induced late response in mild atopic asthma. Am J Respir Cell Mol Biol 1997, 16:664–673.CrossRefPubMed Gizycki MJ, Adelroth E, Rogers AV, O'Byrne PM, Jeffery PK: Myofibroblast involvement in the allergen-induced late response in mild atopic asthma. Am J Respir Cell Mol Biol 1997, 16:664–673.CrossRefPubMed
23.
Zurück zum Zitat Dolhnikoff M, da Silva LFF, de Araujo BB, Gomes HAP, Fernezlian S, Mulder A, Lindeman JH, Mauad T: The outer wall of small airways is a major site of remodeling in fatal asthma. Journal of Allergy and Clinical Immunology 2009,123(5):1090–1097. e1091CrossRefPubMed Dolhnikoff M, da Silva LFF, de Araujo BB, Gomes HAP, Fernezlian S, Mulder A, Lindeman JH, Mauad T: The outer wall of small airways is a major site of remodeling in fatal asthma. Journal of Allergy and Clinical Immunology 2009,123(5):1090–1097. e1091CrossRefPubMed
24.
Zurück zum Zitat Larsen K, Tufvesson E, Malmstrom J, Morgelin M, Wildt M, Andersson A, Lindstrom A, Malmstrom A, Lofdahl CG, Marko-Varga G, et al.: Presence of activated mobile fibroblasts in bronchoalveolar lavage from patients with mild asthma. Am J Respir Crit Care Med 2004, 170:1049–1056.CrossRefPubMed Larsen K, Tufvesson E, Malmstrom J, Morgelin M, Wildt M, Andersson A, Lindstrom A, Malmstrom A, Lofdahl CG, Marko-Varga G, et al.: Presence of activated mobile fibroblasts in bronchoalveolar lavage from patients with mild asthma. Am J Respir Crit Care Med 2004, 170:1049–1056.CrossRefPubMed
25.
Zurück zum Zitat Xisto DG, Farias LL, Ferreira HC, Picanco MR, Amitrano D, Lapa e Silva JR, Negri EM, Mauad T, Carnielli D, Silva LFF, et al.: Lung Parenchyma Remodeling in a Murine Model of Chronic Allergic Inflammation. Am J Respir Crit Care Med 2005,171(8):829–837.CrossRefPubMed Xisto DG, Farias LL, Ferreira HC, Picanco MR, Amitrano D, Lapa e Silva JR, Negri EM, Mauad T, Carnielli D, Silva LFF, et al.: Lung Parenchyma Remodeling in a Murine Model of Chronic Allergic Inflammation. Am J Respir Crit Care Med 2005,171(8):829–837.CrossRefPubMed
26.
Zurück zum Zitat Henderson WR JR, Tang LO, Chu SJ, Tsao SM, Chiang GKS, Jones F, Jonas M, Pae C, Wang H, Chi EY: A Role for Cysteinyl Leukotrienes in Airway Remodeling in a Mouse Asthma Model. Am J Respir Crit Care Med 2002,165(1):108–116.CrossRefPubMed Henderson WR JR, Tang LO, Chu SJ, Tsao SM, Chiang GKS, Jones F, Jonas M, Pae C, Wang H, Chi EY: A Role for Cysteinyl Leukotrienes in Airway Remodeling in a Mouse Asthma Model. Am J Respir Crit Care Med 2002,165(1):108–116.CrossRefPubMed
27.
Zurück zum Zitat Leigh R, Ellis R, Wattie J, Southam DS, De Hoogh M, Gauldie J, O'Byrne PM, Inman MD: Dysfunction and remodeling of the mouse airway persist after resolution of acute allergen-induced airway inflammation. American Journal Of Respiratory Cell And Molecular Biology 2002,27(5):526–535.CrossRefPubMed Leigh R, Ellis R, Wattie J, Southam DS, De Hoogh M, Gauldie J, O'Byrne PM, Inman MD: Dysfunction and remodeling of the mouse airway persist after resolution of acute allergen-induced airway inflammation. American Journal Of Respiratory Cell And Molecular Biology 2002,27(5):526–535.CrossRefPubMed
28.
Zurück zum Zitat Castro-Malaspina H, Gay RE, Resnick G, Kapoor N, Meyers P, Chiarieri D, McKenzie S, Broxmeyer HE, Moore MA: Characterization of human bone marrow fibroblast colony-forming cells (CFU-F) and their progeny. Blood 1980, 56:289–301.PubMed Castro-Malaspina H, Gay RE, Resnick G, Kapoor N, Meyers P, Chiarieri D, McKenzie S, Broxmeyer HE, Moore MA: Characterization of human bone marrow fibroblast colony-forming cells (CFU-F) and their progeny. Blood 1980, 56:289–301.PubMed
29.
Zurück zum Zitat Benjamini Y, Hochberg Y: Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc B 1995, 57:289–300. Benjamini Y, Hochberg Y: Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc B 1995, 57:289–300.
30.
Zurück zum Zitat Sekiya I, Larson BL, Vuoristo JT, Reger RL, Prockop DJ: Comparison of effect of BMP-2, -4, and -6 on in vitro cartilage formation of human adult stem cells from bone marrow stroma. Cell Tissue Res 2005,320(2):269–276.CrossRefPubMed Sekiya I, Larson BL, Vuoristo JT, Reger RL, Prockop DJ: Comparison of effect of BMP-2, -4, and -6 on in vitro cartilage formation of human adult stem cells from bone marrow stroma. Cell Tissue Res 2005,320(2):269–276.CrossRefPubMed
31.
Zurück zum Zitat Gang EJ, Bosnakovski D, Figueiredo CA, Visser JW, Perlingeiro RC: SSEA-4 identifies mesenchymal stem cells from bone marrow. Blood 2007,109(4):1743–1751.CrossRefPubMed Gang EJ, Bosnakovski D, Figueiredo CA, Visser JW, Perlingeiro RC: SSEA-4 identifies mesenchymal stem cells from bone marrow. Blood 2007,109(4):1743–1751.CrossRefPubMed
32.
Zurück zum Zitat McQualter JL, Brouard N, Williams B, Baird BN, Sims-Lucas S, Yuen K, Nilsson SK, Simmons PJ, Bertoncello I: Endogenous fibroblastic progenitor cells in the adult mouse lung are highly enriched in the Sca-1 positive cell fraction. Stem Cells 2008. stemcells.2008–0866 McQualter JL, Brouard N, Williams B, Baird BN, Sims-Lucas S, Yuen K, Nilsson SK, Simmons PJ, Bertoncello I: Endogenous fibroblastic progenitor cells in the adult mouse lung are highly enriched in the Sca-1 positive cell fraction. Stem Cells 2008. stemcells.2008–0866
33.
Zurück zum Zitat Dennis JE, Carbillet JP, Caplan AI, Charbord P: The STRO-1+ marrow cell population is multipotential. Cells Tissues Organs 2002, 170:73–82.CrossRefPubMed Dennis JE, Carbillet JP, Caplan AI, Charbord P: The STRO-1+ marrow cell population is multipotential. Cells Tissues Organs 2002, 170:73–82.CrossRefPubMed
34.
Zurück zum Zitat Blazsek I, Chagraoui J, Péault B: Ontogenic emergence of the hematon, a morphogenetic stromal unit that supports multipotential hematopoietic progenitors in mouse bone marrow. Blood 2000, 96:3763–3771.PubMed Blazsek I, Chagraoui J, Péault B: Ontogenic emergence of the hematon, a morphogenetic stromal unit that supports multipotential hematopoietic progenitors in mouse bone marrow. Blood 2000, 96:3763–3771.PubMed
35.
Zurück zum Zitat Kémoun P, Laurencin-Dalicieux S, Rue J, Vaysse F, Roméas A, Arzate H, Conte-Auriol F, Farges JC, Salles JP, Brunel G: Localization of STRO-1, BMP-2/-3/-7, BMP receptors and phosphorylated Smad-1 during the formation of mouse periodontium. Tissue Cell 2007, 39:257–266.CrossRefPubMed Kémoun P, Laurencin-Dalicieux S, Rue J, Vaysse F, Roméas A, Arzate H, Conte-Auriol F, Farges JC, Salles JP, Brunel G: Localization of STRO-1, BMP-2/-3/-7, BMP receptors and phosphorylated Smad-1 during the formation of mouse periodontium. Tissue Cell 2007, 39:257–266.CrossRefPubMed
36.
Zurück zum Zitat Delorme B, Ringe J, Gallay N, Le Vern Y, Kerboeuf D, Jorgensen C, Rosset P, Sensebé L, Layrolle P, Häupl T, et al.: Specific plasma membrane protein phenotype of culture-amplified and native human bone marrow mesenchymal stem cells. Blood 2008, 111:2631–2635.CrossRefPubMed Delorme B, Ringe J, Gallay N, Le Vern Y, Kerboeuf D, Jorgensen C, Rosset P, Sensebé L, Layrolle P, Häupl T, et al.: Specific plasma membrane protein phenotype of culture-amplified and native human bone marrow mesenchymal stem cells. Blood 2008, 111:2631–2635.CrossRefPubMed
37.
Zurück zum Zitat Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop D, Horwitz E: Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006,8(4):315–317.CrossRefPubMed Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop D, Horwitz E: Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006,8(4):315–317.CrossRefPubMed
38.
Zurück zum Zitat Bentley JK, Deng H, Linn MJ, Lei J, Dokshin GA, Fingar DC, Bitar KN, Henderson WR, Hershenson MB: Airway smooth muscle hyperplasia and hypertrophy correlate with glycogen synthase kinase-3{beta} phosphorylation in a mouse model of asthma. Am J Physiol Lung Cell Mol Physiol 2009, 296:L176–184.CrossRefPubMed Bentley JK, Deng H, Linn MJ, Lei J, Dokshin GA, Fingar DC, Bitar KN, Henderson WR, Hershenson MB: Airway smooth muscle hyperplasia and hypertrophy correlate with glycogen synthase kinase-3{beta} phosphorylation in a mouse model of asthma. Am J Physiol Lung Cell Mol Physiol 2009, 296:L176–184.CrossRefPubMed
39.
Zurück zum Zitat Oh SP, Yeo CY, Lee Y, Schrewe H, Whitman M, Li E: Activin type IIA and IIB receptors mediate Gdf11 signaling in axial vertebral patterning. Genes & Development 2002,16(21):2749–2754.CrossRef Oh SP, Yeo CY, Lee Y, Schrewe H, Whitman M, Li E: Activin type IIA and IIB receptors mediate Gdf11 signaling in axial vertebral patterning. Genes & Development 2002,16(21):2749–2754.CrossRef
40.
Zurück zum Zitat Andersson O, Reissmann E, Ibanez CF: Growth differentiation factor 11 signals through the transforming growth factor-beta receptor ALK5 to regionalize the anterior-posterior axis. EMBO Rep 2006,7(8):831–837.PubMedPubMedCentral Andersson O, Reissmann E, Ibanez CF: Growth differentiation factor 11 signals through the transforming growth factor-beta receptor ALK5 to regionalize the anterior-posterior axis. EMBO Rep 2006,7(8):831–837.PubMedPubMedCentral
41.
Zurück zum Zitat Michos O, Panman L, Vintersten K, Beier K, Zeller R, Zuniga Ae: Gremlin-mediated BMP antagonism induces the epithelial-mesenchymal feedback signaling controlling metanephric kidney and limb organogenesis. Development 2004,131(14):3401–3410.CrossRefPubMed Michos O, Panman L, Vintersten K, Beier K, Zeller R, Zuniga Ae: Gremlin-mediated BMP antagonism induces the epithelial-mesenchymal feedback signaling controlling metanephric kidney and limb organogenesis. Development 2004,131(14):3401–3410.CrossRefPubMed
42.
Zurück zum Zitat Ikeya M, Kawada M, Kiyonari H, Sasai N, Nakao K, Furuta Y, Sasai Y: Essential pro-Bmp roles of crossveinless 2 in mouse organogenesis. Development 2006,133(22):4463–4473.CrossRefPubMed Ikeya M, Kawada M, Kiyonari H, Sasai N, Nakao K, Furuta Y, Sasai Y: Essential pro-Bmp roles of crossveinless 2 in mouse organogenesis. Development 2006,133(22):4463–4473.CrossRefPubMed
43.
Zurück zum Zitat Deng H, Dokshin GA, Lei J, Goldsmith AM, Bitar KN, Fingar DC, Hershenson MB, Bentley JK: Inhibition of Glycogen Synthase Kinase-3{beta} Is Sufficient for Airway Smooth Muscle Hypertrophy. J Biol Chem 2008,283(15):10198–10207.CrossRefPubMedPubMedCentral Deng H, Dokshin GA, Lei J, Goldsmith AM, Bitar KN, Fingar DC, Hershenson MB, Bentley JK: Inhibition of Glycogen Synthase Kinase-3{beta} Is Sufficient for Airway Smooth Muscle Hypertrophy. J Biol Chem 2008,283(15):10198–10207.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Berman JS, Serlin D, Li X, Whitley G, Hayes J, Rishikof DC, Ricupero DA, Liaw L, Goetschkes M, O'Regan AW: Altered bleomycin-induced lung fibrosis in osteopontin-deficient mice. Am J Physiol Lung Cell Mol Physiol 2004,286(6):L1311–1318.CrossRefPubMed Berman JS, Serlin D, Li X, Whitley G, Hayes J, Rishikof DC, Ricupero DA, Liaw L, Goetschkes M, O'Regan AW: Altered bleomycin-induced lung fibrosis in osteopontin-deficient mice. Am J Physiol Lung Cell Mol Physiol 2004,286(6):L1311–1318.CrossRefPubMed
45.
Zurück zum Zitat Kohan M, Breuer R, Berkman N: Osteopontin Induces Airway Remodeling and Lung Fibroblast Activation in a Murine Model of Asthma. Am J Respir Cell Mol Biol 2009,41(3):290–296.CrossRefPubMed Kohan M, Breuer R, Berkman N: Osteopontin Induces Airway Remodeling and Lung Fibroblast Activation in a Murine Model of Asthma. Am J Respir Cell Mol Biol 2009,41(3):290–296.CrossRefPubMed
46.
Zurück zum Zitat Jarvinen L, Badri L, Wettlaufer S, Ohtsuka T, Standiford TJ, Toews GB, Pinsky DJ, Peters-Golden M, Lama VN: Lung Resident Mesenchymal Stem Cells Isolated from Human Lung Allografts Inhibit T Cell Proliferation via a Soluble Mediator. J Immunol 2008,181(6):4389–4396.CrossRefPubMedPubMedCentral Jarvinen L, Badri L, Wettlaufer S, Ohtsuka T, Standiford TJ, Toews GB, Pinsky DJ, Peters-Golden M, Lama VN: Lung Resident Mesenchymal Stem Cells Isolated from Human Lung Allografts Inhibit T Cell Proliferation via a Soluble Mediator. J Immunol 2008,181(6):4389–4396.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Ortiz LA, Gambelli F, McBride C, Gaupp D, Baddoo M, Kaminski N, Phinney DG: Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. PNAS 2003,100(14):8407–8411.CrossRefPubMedPubMedCentral Ortiz LA, Gambelli F, McBride C, Gaupp D, Baddoo M, Kaminski N, Phinney DG: Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. PNAS 2003,100(14):8407–8411.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Rojas M, Xu J, Woods CR, Mora AL, Spears W, Roman R, Brigham KL: Bone marrow-derived mesenchymal stem cells in repair of the injured lung. Am J Respir Cell Mol Biol 2005, 33:145–152.CrossRefPubMedPubMedCentral Rojas M, Xu J, Woods CR, Mora AL, Spears W, Roman R, Brigham KL: Bone marrow-derived mesenchymal stem cells in repair of the injured lung. Am J Respir Cell Mol Biol 2005, 33:145–152.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Ortiz LA, DuTreil M, Fattman C, Pandey AC, Torres G, Go K, Phinney DG: Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proceedings of the National Academy of Sciences 2007,104(26):11002–11007.CrossRef Ortiz LA, DuTreil M, Fattman C, Pandey AC, Torres G, Go K, Phinney DG: Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proceedings of the National Academy of Sciences 2007,104(26):11002–11007.CrossRef
50.
Zurück zum Zitat Gupta N, Su X, Popov B, Lee JW, Serikov V, Matthay MA: Intrapulmonary Delivery of Bone Marrow-Derived Mesenchymal Stem Cells Improves Survival and Attenuates Endotoxin-Induced Acute Lung Injury in Mice. J Immunol 2007,179(3):1855–1863.CrossRefPubMed Gupta N, Su X, Popov B, Lee JW, Serikov V, Matthay MA: Intrapulmonary Delivery of Bone Marrow-Derived Mesenchymal Stem Cells Improves Survival and Attenuates Endotoxin-Induced Acute Lung Injury in Mice. J Immunol 2007,179(3):1855–1863.CrossRefPubMed
51.
Zurück zum Zitat Nilsen-Hamilton MHR, Alvarez-Azaustre E: Relationship between mitogen-regulated protein (MRP) and proliferin (PLF), a member of the prolactin/growth hormone family. Gene 1987,51(2):163–170.CrossRefPubMed Nilsen-Hamilton MHR, Alvarez-Azaustre E: Relationship between mitogen-regulated protein (MRP) and proliferin (PLF), a member of the prolactin/growth hormone family. Gene 1987,51(2):163–170.CrossRefPubMed
52.
Zurück zum Zitat Wiemers DO, Shao Lj, Ain R, Dai G, Soares MJ: The Mouse Prolactin Gene Family Locus. Endocrinology 2003,144(1):313–325.CrossRefPubMed Wiemers DO, Shao Lj, Ain R, Dai G, Soares MJ: The Mouse Prolactin Gene Family Locus. Endocrinology 2003,144(1):313–325.CrossRefPubMed
53.
Zurück zum Zitat Fassett JT, Nilsen-Hamilton M: Mrp3, a Mitogen-Regulated Protein/Proliferin Gene Expressed in Wound Healing and in Hair Follicles. Endocrinology 2001,142(5):2129–2137.PubMed Fassett JT, Nilsen-Hamilton M: Mrp3, a Mitogen-Regulated Protein/Proliferin Gene Expressed in Wound Healing and in Hair Follicles. Endocrinology 2001,142(5):2129–2137.PubMed
54.
Zurück zum Zitat Corbacho A, Martinez De La Escalera G, Clapp C: Roles of prolactin and related members of the prolactin/growth hormone/placental lactogen family in angiogenesis. J Endocrinol 2002,173(2):219–238.CrossRefPubMed Corbacho A, Martinez De La Escalera G, Clapp C: Roles of prolactin and related members of the prolactin/growth hormone/placental lactogen family in angiogenesis. J Endocrinol 2002,173(2):219–238.CrossRefPubMed
55.
Zurück zum Zitat Saha S, Gonzalez J, Rosenfeld G, Keiser H, Peeva E: Prolactin alters the mechanisms of B cell tolerance induction. Arthritis & Rheumatism 2009,60(6):1743–1752.CrossRef Saha S, Gonzalez J, Rosenfeld G, Keiser H, Peeva E: Prolactin alters the mechanisms of B cell tolerance induction. Arthritis & Rheumatism 2009,60(6):1743–1752.CrossRef
Metadaten
Titel
Ovalbumin sensitization and challenge increases the number of lung cells possessing a mesenchymal stromal cell phenotype
verfasst von
J Kelley Bentley
Antonia P Popova
Paul D Bozyk
Marisa J Linn
Amy E Baek
Jing Lei
Adam M Goldsmith
Marc B Hershenson
Publikationsdatum
01.12.2010
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2010
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/1465-9921-11-127

Weitere Artikel der Ausgabe 1/2010

Respiratory Research 1/2010 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Aquatherapie bei Fibromyalgie wirksamer als Trockenübungen

03.05.2024 Fibromyalgiesyndrom Nachrichten

Bewegungs-, Dehnungs- und Entspannungsübungen im Wasser lindern die Beschwerden von Patientinnen mit Fibromyalgie besser als das Üben auf trockenem Land. Das geht aus einer spanisch-brasilianischen Vergleichsstudie hervor.

Wo hapert es noch bei der Umsetzung der POMGAT-Leitlinie?

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

VHF-Ablation nützt wohl nur bei reduzierter Auswurfleistung

02.05.2024 Ablationstherapie Nachrichten

Ob die Katheterablation von Vorhofflimmern bei Patienten mit Herzinsuffizienz die Komplikationsraten senkt, scheint davon abzuhängen, ob die Auswurfleistung erhalten ist oder nicht. Das legen die Ergebnisse einer Metaanalyse nahe.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.