Find information on thousands of medical conditions and prescription drugs.

Bronchopulmonary dysplasia

Bronchopulmonary dysplasia (BPS) is a chronic lung disorder that is most common among children who were born prematurely, with low birthweights and who received prolonged mechanical ventilation to treat respiratory distress syndrome. It is characterized by inflammation and scarring in the lungs. It develops most commonly in the first 4 weeks after birth.

Home
Diseases
A
B
Babesiosis
Bacterial endocarditis
Bacterial food poisoning
Bacterial meningitis
Bacterial pneumonia
Balantidiasis
Bangstad syndrome
Bardet-Biedl syndrome
Bardet-Biedl syndrome
Bardet-Biedl syndrome
Bardet-Biedl syndrome
Barrett syndrome
Barth syndrome
Basal cell carcinoma
Bathophobia
Batrachophobia
Batten disease
Becker's muscular dystrophy
Becker's nevus
Behcet syndrome
Behr syndrome
Bejel
Bell's palsy
Benign congenital hypotonia
Benign essential tremor...
Benign fasciculation...
Benign paroxysmal...
Berdon syndrome
Berger disease
Beriberi
Berylliosis
Besnier-Boeck-Schaumann...
Bibliophobia
Bicuspid aortic valve
Biliary atresia
Binswanger's disease
Biotinidase deficiency
Bipolar disorder
Birt-Hogg-Dube syndrome
Blastoma
Blastomycosis
Blepharitis
Blepharospasm
Bloom syndrome
Blue diaper syndrome
Blue rubber bleb nevus
Body dysmorphic disorder
Boil
Borreliosis
Botulism
Bourneville's disease
Bowen's disease
Brachydactyly
Brachydactyly type a1
Bradykinesia
Bright's disease
Brittle bone disease
Bromidrosiphobia
Bronchiectasis
Bronchiolotis obliterans...
Bronchopulmonary dysplasia
Brown-Sequard syndrome
Brucellosis
Brugada syndrome
Bubonic plague
Budd-Chiari syndrome
Buerger's disease
Bulimia nervosa
Bullous pemphigoid
Burkitt's lymphoma
Byssinosis
Cavernous angioma
C
D
E
F
G
H
I
J
K
L
M
N
O
P
Q
R
S
T
U
V
W
X
Y
Z
Medicines

Read more at Wikipedia.org


[List your site here Free!]


Bombesin-like peptides and mast cell responses: Relevance to bronchopulmonary dysplasia?
From American Journal of Respiratory and Critical Care Medicine, 9/1/03 by Subramaniam, Meera

Bombesin-like peptides (BLPs) are elevated in newborns who later develop bronchopulmonary dysplasia (BPD). In baboon models, anti-BLP blocking antibodies abrogate BPD. We now demonstrate hyperplasia of both neuroendocrine cells and mast cells in lungs of baboons with BPD, compared with non-BPD controls or BLP antibody-treated BPD baboons. To determine whether BLPs are proinflammatory, bombesin was administered intratracheally to mice. Forty-eight hours later, we observed increased numbers of lung mast cells. We analyzed murine mast cells for BLP receptor gene expression, and identified mRNAs encoding bombesin receptor subtype 3 and neuromedin-B receptor (NMB-R), but not gastrin-releasing peptide receptor. Only NMB-R-null mice accumulated fewer lung mast cells after bombesin treatment. Bombesin, gastrin-releasing peptide, NMB, and a bombesin receptor subtype 3-specific ligand induced mast cell proliferation and chemotaxis in vitro. These observations support a role for multiple BLPs in promoting mast cell responses, suggesting a mechanistic link between BLPs and chronic inflammatory lung diseases.

Keywords: bronchopulmonary dysplasia; chemotaxis; infant, premature; neuromedin B; pulmonary fibrosis

Bombesin is a 14-amino acid bioactive peptide originally isolated from frog skin (1). Bombesin-like peptides (BLPs) are mammalian homologs of bombesin that are elevated in newborns who later develop bronchopulmonary dysplasia (BPD) (2). BLPs comprise a family of related peptides, of which several could account for BLP immunoreactivity localized to the pulmonary neuroendocrine (NE) cells, with highest BLP levels occurring in fetal lung (3). One major pulmonary BLP was later identified as gastrin-releasing peptide (GRP) (1). GRP and bombesin are highly similar both structurally and functionally, and thus are collectively referred to as "BLPs"; neuromedin B (NMB) is also closely related to bombesin (1, 4). BLPs are growth factors for 3T3 fibroblasts, normal airway epithelial cells, and many cancer cell lines (1, 5); have neuroregulatory functions in the brain and gut; and act as potent bronchoconstrictors (6). Multiple receptor subtypes mediate effects of bombesin-related peptides, and three mammalian genes for these G protein-coupled receptors have been cloned: the GRP/bombesin (BLP)-preferring receptor (GRP-R), the neuromedin B receptor (NMB-R), and the orphan bombesin receptor subtype-3 (BRS-3), for which no natural ligand is known (7). A related receptor has been cloned in the frog and is known as bombesin receptor subtype-4 (8). In fetal lung, GRP-R mRNA is localized predominantly in the undifferentiated mesenchyme around developing airways and blood vessels, with lower levels in airway epithelium (9). GRP-R, NMB-R, and BRS-3 have also been identified in many human cancers and cancer cell lines and some corresponding normal cell types (1, 10-15).

In previous studies, we demonstrated that BLPs promote fetal lung development, including lung branching morphogenesis, epithelial and mesenchymal cell proliferation, and Type II cell differentiation (1). GRP and GRP-R mRNAs fall to undetectable levels after birth but may be upregulated poslnalally in premature baboons with bronchopulmonary dysplasia (BPD, also known as chronic lung disease of prematurity or CLD) (16). Increased numbers of BLP-positive NE cells occur in lungs of infants dying of BPD (17, 18). Elevated levels of BLPs and/or increased numbers of BLP-positive NE cells have been associated with other chronic inflammatory lung diseases (1), including cystic fibrosis, lung cancer, eosinophilic granuloma, tobacco-related lung disorders, and pulmonary hypertension. A few previous reports suggested a possible role for BLPs in promoting cellular responses that may contribute to inflammatory processes in vitro, including macrophage activation and phagocytosis (1), chemotaxis of macrophages and fibroblasts (1, 19, 20), smooth muscle constriction (21), and proliferation of fibroblasts, lymphocytes, and endothelial cells (20, 22, 23). In spite of all these observations, no mechanistic link has been established between BLPs and pulmonary inflammation in vivo.

BPD is the most common chronic lung disease in infants in the United States (24, 25). Recognized contributing factors include mechanical ventilation, oxygen toxicity, infection, and pulmonary immaturity. Nonetheless, the pathophysiology of BPD remains enigmatic. It is unclear why only a subset of premature infants develops BPD (24). Two different premature baboon models of BPD have been valuable tools for investigating the pathophysiology of this disorder (26, 27): "hyperoxic" animals gestated for 140 days (140d/100%), and "interrupted gestation" (125-day) animals given maintenance levels of oxygen (125d/PRN [pro re nata, as needed]). Previously (18), we observed elevated urine BLP levels in both of these baboon models of BPD. Administration of anti-BLP blocking antibody 2A11 to baboons in either model abrogates the lung injury characteristic of BPD (18). We demonstrated elevated BLPs shortly after birth in premature human infants who later develop BPD (28). Cumulatively, these observations prompted us to test the hypothesis that BLPs function postnatally as proinflammatory cytokines. In the present study, we focus our analysis on mast cells, which are present in diverse inflammatory lung diseases.

METHODS

Animals

Mice housed at Children's Hospital Boston (Boston, MA) included 5- to 6-week-old Swiss-Webster females (Taconic Farms, Germantown, NY) and GRP-R-null, NMB-R-null, and BRS-3-null mice (29-31). Protocols were approved by the Institutional Animal Care and Use Committee of Children's Hospital Boston.

Baboons were studied at the Southwest Foundation for Biomedical Research (San Antonio, TX) as described (16, 18).

Premature baboons from the hypcroxic model of BPD (140/100% X 10d) (26) were given anti-BLP antibody (2A11) or the control IgG1, MOPC21 (18). On Day 10, right middle lobes inflated with 4% paraformaldehyde were fixed overnight. Sections chosen included complete lung cross-sections from proximal (cartilaginous) conducting airways down to the distal parenchyma (alveoli and pleura).

Intratracheal Bombesin

Seventy-five [mu]l of sterile phosphate-buffered saline (PBS)-1% bovine serum albumin (BSA)-0.5% Evans blue was instilled into anesthetized mice via a transoral/intratracheal catheter with or without bombesin (5 [mu]g). There was no mortality of the mice in any experimental groups. After CO^sub 2^ euthanasia, lungs were inflated in situ with 4% paraformaldehyde.

Histochemistry

Five-micrometer baboon lung paraffin sections were immunostained for tryplase with monoclonal antibody clone AA1 and the Animal Research Kit peroxidase system (Dako Laboratories, Carpinteria, CA). BLP immunostaining is described elsewhere (18). Alcian blue-safranin histochemistry was performed to detect murine mast cells (32).

Morphometry

Positive cells (NE cells or mast cells) were counted across entire slides. Tissue section areas were determined by computer-assisted image analysis (Scion Image, version 1.62b; Scion, Frederick, MD) and results were expressed as number of cells per unit area (square millimeters for baboons, square centimeters for mice) (33).

Cultured Mast Cell Line

MC/9 cells were cultured as described (American Type Culture Collection, Manassas, VA) (34, 35). This cloned normal murine mast cell line bears IgE receptors and produces histaminc and leukotrienes.

Bone Marrow-derived Mast Cells

Bone marrow cells from Swiss-Webster mice were cultured as described (36). Purity was assessed by flow cytometry for surface IgE (36).

Reverse Transcription-Polymerase Chain Reaction

Reverse transcription-polymerase chain reaction (RT-PCR) was performed as described (37): 25 cycles for glyceraldehyde-3-phosphate dehydrogenase and 35 cycles for GRP-R, NMB-R, and BRS-3 (4, 38, 39). A second round of PCR (10 cycles) used nested primers for GRP-R, BRS-3, and NMB-R. Glyceraldehyde-3-phosphate dehydrogenase primers were from BD Biosciences Clontech (Palo Alto, CA). PCR primers for murine GRP-R, NMB-R, and BRS3 are described in the online supplement.

Mast Cell Proliferation

MC/9 cells were cultured at 37[degrees]C in 5% CO^sub 2^ and serum-free Dulbecco's modified Eagle's medium (DMEM)-0.1% BSA (DMEM-BSA) for 18 hours before assay. Cells (15 X 10^sup 4^/ml) were plated in 6-well plates with DMEM-BSA with or without 0.1-100 nM bombesin (NMB; Peninsula Laboratories, San Carlos, CA), or with BRS-3-specific synthetic peptide 3209 [D-, Tyr^sup 6^,[beta]-Ala^sup 11^,Phe13,Nle^sup 14^]bombesin^sub (6-14)^ (BRS-3L) (40). After 24 hours, cells were harvested for Ki-67 flow cytometry (41, 42). GRP-R-specific antagonist D-Tyr^sup 6^-bombesin^sub (6-13)^ methyl ester (GRA) was also used (43). No ligand or antagonist had any effect on cell viability, as determined by trypan blue exclusion.

Chemotaxis

MC/9 cells (10^sup 6^ in 100 [mu]l of PBS-BSA) were plated in the upper chambers of Transwell inserts (pore size, 5 [mu]m; 6.5 mm in diameter; polycarbonate) in 24-well plates and incubated for 3 hours at 37[degrees]C. The lower chambers had 600 [mu]l of PBS-BSA with or without 0.1-10 nM bombesin, GRP^sub (1-27)^, GRP^sub (1-14)^, GRP^sub (16-27)^, NMB (Peninsula Laboratories), or BRS-3L (40). Upper chamber cells were sometimes treated with GRA (43). Lower chamber cells were counted with a hemacytomeler. Numbers were expressed as a percentage of total number of cells added to the upper wells.

Statistics

For statistical analyses we used an unpaired Student / test or analysis of variance.

RESULTS

Pulmonary NE Cells in BPD and Non-BPD Baboons

Increased urine BLP levels are known to occur in the hyperoxic preterm baboon model of BPD (140d/100% baboons) (18). One of the main sources of this elevated BLP immunoreactivity is believed to be increased BLP production by pulmonary NE cells. To lest this hypothesis, we performed morphometric analyses of BLP-positive cells in lung sections from these baboons. Similar to our earlier studies of hyperoxic hamsters, there was an increase in both the total number of clusters (neuroepithelial body-like foci) and in the number of nucleated cells per cluster. Note that by definition, neuroepithelial bodies must be innervated and this could not be evaluated on the given slides because pulmonary nerve fibers are negative for BLP immunostaining. NE cells staining for the NE/neural cytoplasmic marker PGP9.5 were increased, similarly to BLP-positive cells, but abundant PGP9.5-positive mesenchymal cells obscured evaluation of nerve fibers (M. Sunday, data not shown). We quantified the total number of BLP-positive pulmonary neuroendocrine cells (PNECs) per unit area of lung to take into account both the increased number of clusters and the increased number of cells per cluster. Representative photomicrographs are given in Figures 1a-1c. The pooled results from morphometric analyses are summarized in Figure 1d. Compared with non-BPD baboons treated for 10 days with O^sub 2^ PRN (pro re nata, sufficient to maintain hemoglobin saturation at about 90%) (Figures 1a and 1d), there was an overall ninefold increase in BLP-positive NE cells in 140d/100% baboons with BPD (Figures 1b and 1d) (p

Mast Cells in Lungs of Baboons with BPD

Considering one report of mast cell hyperplasia in lungs of human infants with BPD (44), we quantitated the numbers of lung mast cells occurring in hyperoxic baboons with BPD (141d/ 100%, BPD group). These results were compared with those of the 140d/PRN, non-BPD group and with 140d/100% baboons treated with anti-BLP antibody 2A11. We determined that immunostaining for human tryptase is the most sensitive and specific method for detecting mast cells in baboon lung (Figures 2a-2c). Similar results were observed when mast cells were stained red, using chloroacetate esterase histochemistry (Figure 2d). Tryptase-positive mast cells were quantified in lung sections by two types of computerized image analysis: (1) as total mast cells per square millimeter of lung tissue (Figure 2e), and (2) as mast cells present in the distal lung parenchyma (i.e., interstitial mast cells, excluding those associated with blood vessels and conducting airways). The total number of mast cells was increased fourfold in the 140d/100% group (14.7/mm^sup 2^ lung tissue) compared with 140d/PRN baboons (3.6/mm^sup 2^) (p

Lung Mast Cells in Mice Given BLP Intratracheally

To directly test the hypothesis that BLPs can function as proinfiammatory cytokines in the lung, we administered bombesin (5 [mu]g in 75 [mu]l of PBS-BSA) or vehicle alone (PBS-BSA) intratracheally to anesthetized mice. Lungs were harvested after 4 hours, 48 hours, 7 days, or 14 days and paraffin-embedded sections were stained with Alcian blue-safranin (32) to demonstrate mast cells, which we determined to be the most sensitive method for detection of these cells in mouse lung (Figures 3a and 3b). The relative density of mast cells was lower in lung sections from mice given a single BLP treatment as compared with sections from BPD baboons treated with hyperoxia for more than 1 week. We observed a twofold increase in numbers of mast cells per square centimeter of lung tissue 48 hours and 7 days after bombesin administration (Figures 3a and 3c) as compared with mice given saline vehicle alone (Figure 3b) (p

To determine which of the three cloned mammalian BLP receptors might mediate these mast cell responses, we administered bombesin intratracheally (5 [mu]g/mouse) to mice genetically deficient for GRP-R, NMB-R, or BRS-3, and their normal littermates, keeping in mind the caveat that such mice can have altered expression of other cytokine/mediator-signaling pathways. Knockout mice were used rather than BLP receptor-specific antagonists because most antagonists are not entirely specific for only one BLP receptor subtype and partial agonist effects are common. The only exception is the GRP-R-specific antagonist (D-Tyr^sup 6^-bombesin^sub (6-13)^ methyl ester, or GRA), which we were able to obtain in only small quantities that precluded its use for in vivo blocking studies. Furthermore, the use of the BLP blocking antibody 2A11 would not give us any pharmacologic information about the receptors involved, but would simply confirm that 2A11 binds BLPs sharing the same C-terminal 7-amino acid sequence as bombesin and GRP.

Only experiments with NMB-R-null mice resulted in 50% fewer mast cells accumulating in lung tissues, compared with wild-type littermates at 48 hours (Figure 3e). At baseline, there was no significant difference in mast cell numbers between NMB-R-null and their wild-type littermates. There was no difference in the numbers of BLP-induced mast cells between wild-type littermates and GRP-R-null or BRS-3-null mice. We did not see a significant difference in the number of mast cells at baseline in any of the untreated mice (data not shown).

GRP-R, NMB-R, and BRS-3 Gene Expression by Mast Cells

To determine whether bombesin might directly trigger murine mast cell responses, we analyzed gene expression for BLP receptors in MC/9, a cloned mouse mast cell line, and in murine bone marrow-derived mast cells (more than 99% pure by flow cytometry and more than 99% viable by trypan blue exclusion). Semiquantitative RT-PCR was performed to assess relative levels of the mRNAs encoding the three BLP receptors: GRP-R, NMB-R, and BRS-3. NMB-R and BRS-3 mRNAs were detected in both types of mast cells (Figures 4A and 4B). GRP-R mRNA was not detected by RT-PCR even when nested primers were used to reamplify the first RT-PCR products in a second round of PCR for up to 10 cycles. The identities of the PCR products were confirmed by sequencing.

BLP-induced Proliferation of Mast Cells

We hypothesized that cell proliferation is one mechanism by which bombesin could increase mast cell numbers in mouse lung. Also, NMB is known to act as a growth factor for lung cancer cell lines (46). The cloned mast cell line MC/9 was chosen for our studies because the numbers of bone marrow-derived mast cells was insufficient for carrying out controlled experiments with samples in duplicate or triplicate to ensure reproducibility. Mast cell proliferation induced by several different BLP ligands was evaluated by flow cytometry for Ki-67, a well characterized marker of proliferating cells. Significantly increased Ki-67 labeling was observed with bombesin at 10 nM (1.7-fold increase over baseline, p

To determine whether NMB-R and/or BRS-3 could be implicated in the MC/9 proliferative response, we tested two other ligands. NMB induced a 1.4-fold increase in MC/9 proliferation at both 1.0 and 10 nM (p

Mast Cell Chemotaxis in Response to BLPs

Increased mast cells in the lungs of BPD baboons were observed primarily in the distal lung parenchyma, whereas control non-BPD baboons had a more proximal distribution of these cells. We tested the ability of BLPs to induce mast cell (MC/9) chemotaxis. MC/9 chemotaxis was significantly increased in the presence of bombesin (2.5- to 3.5-fold increase at 1 and 10 nM bombesin, respectively; p

DISCUSSION

The present study demonstrates two major lines of evidence supporting a previously missing direct link between BLPs and mast cell hyperplasia. First, in lungs of hyperoxic premature baboons, endogenous BLPs mediate hyperplasia of NE cells and mast cells: anti-BLP blocking antibody (2A11) abrogates these increases. Previously, we showed that 2A11 treatment abrogates the development of BPD in these same hyperoxic hyperoxic baboons, both clinically and pathologically (18). Second, in mice, intratracheal bombesin induces lung mast cell accumulation. Cumulatively, these observations suggest that BLPs could play a role in the pathophysiology of chronic inflammatory lung diseases by triggering a cellular cascade including mast cells.

Hyperplastic pulmonary NE cells are likely to be the source of elevated urine BLP levels in newborns with BPD (18, 28). This hyperplasia could be induced by oxidant injury (47), tumor necrosis factor-[alpha] (49), or other cytokines, including BLPs (50, 51). It is known that PNECs in normal lung occur both as isolated cells (mainly in large airways) and as small clusters called neuroepithelial bodies that are concentrated at branch points within the epithelium of branching airways. Hyperplastic PNECs induced by hyperoxia, nitrosamine carcinogens, cigarette smoking, or in idiopathic primary PNEC hyperplasia are often preferentially localized to the small bronchioles and alveolar ducts (2, 18, 52-55). Similarly, there are also more mast cells and other inflammatory infiltrates in the small airways in patients with nonfatal asthma or status asthmaticus (56, 57). Cumulatively, these observations suggest that NE cells can function as proinflammatory cells.

BLPs could promote lung remodeling in BPD by triggering proliferation of both epithelial and mesenchymal cells (58). BLP levels are increased shortly after birth in infants who later develop BPD (18, 28). Mast cells can function in both acute and chronic inflammation, promoting both innate and acquired immune responses including leukocyte recruitment by chemotaxis (59, 60), and leading to lung injury and fibrosis in later stages (61-63). The current study demonstrates increased mast cells in the lung in response to BLPs. This inflammatory response may have a role in the pathogenesis of BPD. Proinflammatory cytokines, adhesion molecules, and inflammatory cells including mast cells have been demonstrated in the lung parenchyma of infants with BPD (44, 64, 65). The concept that BLP-induced mast cell responses might be an early event leading to lung injury in BPD represents a paradigm shift in current understandings of BPD. These observations could be of further relevance because BPD is associated with bronchospasm and inflammation, similar to asthma (61, 62).

BLP is well known as a growth factor and neuroregulatory peptide (1, 22, 23). However, no prior study has documented gene expression for BLP receptors in hematopoietic cells, in particular mast cells. Two mechanisms by which BLPs could increase mast cells in the lung are by directly triggering mast cell proliferation and/or chemotaxis. We demonstrate that two of the three genes encoding mammalian BLP receptors, BRS-3 and NMB-R, are expressed by murine mast cells (MC/9 and bone marrow-derived mast cells). The functional relevance of this receptor gene expression is supported by the ability of BRS-3-and NMB-R-specific ligands to elicit significant mast cell proliferation and chemotaxis at low doses (1.0 nM or less). Proliferation induced by 10 nM bombesin is probably mediated via NMB-R on mast cells (66), because bombesin has lower affinity for NMB-R compared with NMB. Bombesin has low affinity for BRS-3 in pharmacologic studies (67). It is unlikely that the GRP-R is involved in bombesin-induced mast cell responses because GRP-R mRNA is undetectable in mast cells, even using nested RT-PCR. Furthermore, a GRP-R-specific antagonist (GRA) (42, 68) does not significantly inhibit bombesin-induced mast cell proliferation or chemotaxis. The observation that mast cell responses can be elicited by both bombesin and GRP, but not by inactive GRP analogs, suggests the involvement of additional bombesin/GRP-preferring receptors, such as a phyllolitorin receptor (50, 67) or a homolog of the frog BB4 receptor (8). Previously, NMB-R and GRP-R have been implicated in cell proliferation in vitro (69, 70), whereas BRS-3 signaling increases calcium mobilization and metabolism but not mitogenesis in cancer cell lines (14). Our observed stimulation of mast cell proliferation using a BRS-3-specific ligand represents a novel function for this BLP receptor subtype, and suggests a role for BRS-3 in inflammatory responses.

Functional studies in genetically deficient mice indicate that null mutations of the three cloned murine BLP receptors yield distinct phenotypes at baseline (29-31). However, none of the previous studies addressed the effects of BLPs in BLP receptor-null mice. We now observe that NMB-R-null mice treated with bombesin have significantly less mast cell hyperplasia compared with wild-type littermates, despite there being no difference in mast cell numbers or distribution at baseline. The lack of such a difference between wild-type controls and GRP-R or BRS-3-null mice is logical: murine mast cells lack GRP-R, and bombesin has low affinity for BRS-3 receptors.

In hyperoxic baboons with BPD, we observed the greatest number of mast cells in the lung parenchyma (alveolar interstitium) distal to the conducting airways. Normally, lung mast cells occur around blood vessels, large airways, and nerves. Others have noted a similar distribution of tryptase-positive mast cells in lungs of human infants dying of BPD (44). BLP-positive NE cells are also found predominantly in the small bronchioles and alveolar ducts in infants with BPD (2, 18). The localization of mast cells to the distal lung parenchyma in infants with BPD may be due to mast cell migration in response to high local concentrations of BLPs. Similarly, mice given bombesin intratracheally have a higher density of mast cells in the lung parenchyma than in the whole lung. We demonstrate chemotaxis of mast cells in response to BLPs, similar to macrophages, epithelial cells, and fibroblasts (1, 20). Cumulatively, these data suggest that BLPs can promote increased parenchymal mast cells in BPD, which in turn may be involved in promoting the chronic inflammation and interstitial fibrosis characteristic of BPD. At present, we cannot rule out additional mechanisms for BLP-elicited accumulation of mast cells.

Genetic variation could explain why only some premature infants develop BPD. A family history of asthma is associated with a 12-fold increased risk of BPD (72, 73). Similarly, BLPs may be implicated in the pathophysiology of asthma, either as direct bronchoconstrictors (6) and/or by modulating the numbers of mast cells in the lung. Our present observations suggest a possible link between NE cells and the pathophysiology of asthma. There have been several reports of NE cell hyperplasia in animals sensitized for allergic airway reactions, with NE cell degranulation occurring after specific antigen challenge (1). Abnormal NE cells and/or NE cell hyperplasia have been identified in lungs of infants exposed to smoke in utero; such children of mothers who smoke are at a threefold increased risk of developing pediatric asthma (1). Finally, many of the bioactive peptides produced by pulmonary NE cells are potent bronchoconstrictors (1). The present study indicates that NE cell-derived peptides may have multiple roles in promoting airway inflammatory responses.

In conclusion, hyperoxic lung injury in premature baboons can be mediated by excessive production of BLPs (18), apparently derived from hyperplastic BLP-positive pulmonary NE cells. Cellular mechanisms for this injury include a cascade leading from NE cells to mast cells, via BRS-3 and NMB receptors on mast cells. Bronchoconstriction can occur early in the course of BPD (74), possibly secondary to mast cell accumulation elicited by BLPs. The definitive significance of mast cells for the pathogenesis of BPD in hyperoxic baboons remains to be clarified. As part of the innate immune system, lung mast cells can promote both innate and acquired immune responses by production of cytokines leading to inflammation, injury, and pulmonary fibrosis in later stages (61-63). Finally, the role of mast cells in the pathogenesis of "new BPD" that is currently observed in very low birthweight infants (24) is unclear but remains under investigation in the extremely premature baboon model of BPD (27). In conclusion, the current study opens new avenues for investigation of the pathophysiology of chronic inflammatory lung diseases.

Conflict of Interest Statement: M.S. has no declared conflict of interest; K.S. has no declared conflict of interest; D.H.C. has no declared conflict of interest; Y.K. has no declared conflict of interest; Y.E.M. has no declared conflict of interest; P. F. W. has no declared conflict of interest; K.W. has no declared conflict of interest; E.W. has no declared conflict of interest; M.E.S. has no declared conflict of interest.

Acknowledgment: The authors thank the NIH for support of the Collaborative Program in BPD, headed by Dr. Jacqueline Coalson, including Dr. Brad Yoder, the Department of Pathology staff at the University of Texas Health Sciences Center (San Antonio, TX), the NICU and animal production staffs at the Southwest Foundation for Biomedical Research (San Antonio, TX), and Vicki Winter. The authors also thank Dr. James Crapo for helpful discussions.

References

1. Sunday ME. Neuropeptides and lung development. In: McDonald JA, editor. Lung growth and development. New York: Marcel Dekker; 1997. p. 401-494.

2. Johnson DE, Lock JE, Elde RP, Thompson TR. Pulmonary neuroendocrine cells in hyaline membrane disease and bronchopulmonary dysplasia. Pediatr Res 1982:16:446-454.

3. Wharton J, Polak JM, Bloom SR, Ghatei MA, Solcia E, Brown MR, Pearse AGE. Bombesin-like immunoreactivily in the lung. Nature 1978; 273:769-770.

4. Wada E, Way J, Shapira H, Kusano K, Lebacq-Verheyden AM, Coy D, Jensen R, Battey J. cDNA cloning, characterization, and brain region-specific expression of a neuromedin-B-preferring bombesin receptor. Neuron 1991;6:421-430.

5. Siegfried JM, Guentert PJ, Gaither AL. Effects of bombesin and gastrin-releasing peptide on human bronchial epithelial cells from a series of donors: individual variation and modulation by bombesin analogs. Anat Rec 1993;236:241-247.

6. Impicciatore M, Bertaccini G. The bronchoconstriclor action of the tetradecapeptide bombesin in the guinea-pig. J Pharm Pharmacol 1973;25:872-875.

7. Kroog GS, Jensen RT, Battey JF. Mammalian bombesin receptors. Med Res Rev 1995;15:389-417.

8. Nagalla SR, Barry BJ, Creswick KC, Eden P, Taylor JT, Spindel ER. Cloning of a receptor for amphibian [Phe13]bombesin distinct from the receptor for gastrin-releasing peptide: identification of a fourth bombesin receptor subtype (BB4). Proc Natl Acad Sci USA 1995;92:6205-6209.

9. Sunday ME. Bioactive peptides and lung development. In: Gaultier C, Bourbon JR, Post M, editors. Lung development. New York: Oxford University Press; 1999. p. 304-326.

10. Toi-Scott M, Jones CL, Kane MA. Clinical correlates of bombesin-like peptide receptor subtype expression in human lung cancer cells. Lung Cancer 1996;15:341-354.

11. Kiaris H, Schally AV, Nagy A, Sun B, Armatis P, Szepeshazi K. Targeted cytotoxic analogue of bombesin/gastrin-releasing peptide inhibits the growth of H-69 human small-cell lung carcinoma in nude mice. Br J Cancer 2001;81:966-971.

12. Sun B, Schally AV, Halmos G. The presence of receptors for bombesin/ GRP and mRNA for three receptor subtypes in human ovarian epithelial cancers. Regul Pept 2000;90:77-84.

13. Sun B, Halmos G, Schally AV, Wang X, Martinez M. Presence of receptors for bombesin/gastrin-releasing peptide and mRNA for three receptor subtypes in human prostate cancers. Prostate 2000;42:295-303.

14. Ryan RR, Weber HC, Mantey SA, Hou W, Hilburger ME, Pradhan TK, Coy DH, Jensen RT. Pharmacology and intracellular signaling mechanisms of the native human orphan receptor BRS-3 in lung cancer cells. J Pharmacol Exp Ther 1998;287:366-380.

15. DeMichele MA, Davis AL, Hunt JD, Landrencau RJ, Siegfried JM. Expression of mRNA for three bombesin receptor subtypes in human bronchial epithelial cells. Am J Respir Cell Mol Biol 1994;11:66-74.

16. Cullen A, Emanuel RL, Haley KJ, Torday JS, Asokananthan N, Sikorski KA, Sunday ME. Bombesin-like peptide (BLP) and BLP receptors in two different baboon models of bronchopulmonary dysplasia: diversity in gene expression yet similarity in function. Peptides 2000;21:1627-1638.

17. Johnson DE, Anderson WR, Burke BA. Pulmonary neuroendocrine cells in pediatric lung disease: alterations in airway structure in infants with bronchopulmonary dysplasia. Anat Rec 1993;236:115-119.

18. Sunday ME, Yoder BA, Cuttitta F, Haley KJ, Emanuel RL. Bombesin-like peptide mediates lung injury in a baboon model of bronchopulmonary dysplasia. J Clin Invest 1998;102:584-594.

19. Kim JS, McKinnis VS, White SR. Migration of guinea pig airway epithelial cells in response to bombesin analogues. Am J Respir Cell Mol BM 1997;16:259-266.

20. Yule KA, White SR. Migration of 3T3 and lung fibroblasts in response to calcitonin gene-related peptide and bombesin. Exp Lung Res 1999; 25:261-273.

21. Erspamer GF, Mazzanti G, Farruggia G, Nakajima T, Yanaihara N. Parallel bioassay of litorin and phyllolitorins on smooth muscle preparations. Peptides 1984;5:765-768.

22. Del Rio M, Hernanz A, De la Fuente M. Bombesin, gastrin-releasing peptide, and neuromedin C modulate murine lymphocyte proliferation through adherent accessory cells and activate protein kinase C. Peptides 1994;15:15-22.

23. Marion-Audibert AM, Nejjari M, Pourreyron C, Anderson W, Gouysse G, Jacquier MF, Dumortier J, Scoazec JY. Effects of endocrine peptides on proliferation, migration and differentiation of human endothelial cells [in French].Gastroenterol Clin Biol 2000;24:644-648.

24. Northway WH. Bronchopulmonary dysplasia: twenty-five years later. Pediatrics 1992;89:969-973.

25. Miller VL, Rice JC, DeVoe M, Fos PJ. An analysis of program and family costs of managed care for technology-dependent infants with bronchopulmonary dysplasia. J Pediatr Nurs 1998;13:244-251.

26. Coalson JJ, Kuehl TJ, Escobedo MB, Hilliard JL, Smith F, Meredith K, Null DM, Walsh W, Johnson D, Robotham JL. A baboon model of bronchopulmonary dysplasia. II. Pathologic features. Exp Mol Pathol 1982;37:335-350.

27. Coalson JJ, Winter VT, Siler-Khodr T, Yoder BA. Neonatal chronic lung disease in extremely immature baboons. Am J Respir Crit Care Med 1999;160:1333-1346.

28. Cullen A, Van Marter LJ, Moore M, Parad R, Sunday ME. Urine bombesin-like peptide elevation precedes clinical evidence of bronchopulmonary dysplasia. Am J Respir Crit Care Med 2002;165:1093-1097.

29. Wada E, Watase K, Yamada K, Ogura H, Yamano M, Inomata Y, Eguchi J, Yamamoto K, Sunday ME, Maeno H, et al. Generation and characterization of mice lacking gastrin-releasing peptide receptor. Biochem Biophys Res Commun 1997;239:28-33.

30. Ohki-Hamazaki H, Watase K, Yamamoto K, Ogura H, Yamano M, Yamada K, Maeno H, Imaki J, Kikujama S, Wada E, et al. Mice lacking bombesin receptor subtype-3 develop metabolic defects and obesity. Nature 1997;390:165-169.

31. Ohki-Hamazaki H, Sakai Y, Kamata K, Ogura H, Okuyama S, Watase K, Yamada K, Wada K. Functional properties of two bombesin-like peptide receptors revealed by the analysis of mice lacking neuromedin B receptor. J Neurosc 1999;19:948-954.

32. Culling CFA, Allison RT, Barr WT. Cellular pathology technique. London: Butterworths; 1985.

33. Whimster WF. Diagnostic morphometry in emphysema and chronic bronchitis. Anal Quant Cytol Histol 1985;7:183-186.

34. Ito F, Toyota N, Sakai H, Takahashi H, Iizuka H. FK506 and cyclosporin A inhibit stem cell factor-dependent cell proliferation/survival, while inducing upregulation of c-kit expression in cells of the line MC/9. Arch Dermatol Res 1999;291:275-283.

35. Bryant RW, She HS, Ng KJ, Siegel MI. Modulation of the 5-lipoxygenase activity of MC-9 mast cells: activation by hydroperoxides. Prostaglandins 1986;32:615-627.

36. Pivniouk VL, Martin TR, Lu-Kuo JM, Katz HR, Oettgen HC, Geha RS. SLP-76 deficiency impairs signaling via the high-affinity IgE receptor in mast cells. J Clin Invest 1999;103:1737-1743.

37. Willett CG, Smith DI, Shridhar V, Wang M-H, Emanuel RL, Patidar K, Graham SA, Zhang F, Hatch V, Sugarbaker DJ, et al. Differential screening of a human chromosome 3 library identifies hepatocyte growth factor-like/macrophage-stimulating protein and its receptor in injured lung: possible implications for neuroendocrine cell survival. J Clin Invest 1997;99:2979-2991.

38. Battey JF, Way JM, Corjay MH, Shapira H, Kusano K, Harkins R, Wu JM, Slattery T, Mann E, Feldman RI. Molecular cloning of the bombesin/gastrin-releasing peptide receptor from Swiss 3T3 cells. Proc Natl Acad Sci USA 1991;88:395-399.

39. Fathi Z, Corjay MH, Shapira H, Wada E, Benya R, Jensen R, Viallet J, Sausville EA, Battey JF. BRS-3: a novel bombesin receptor subtype selectively expressed in testis and lung carcinoma cells. J Biol Chem 1993;268:5979-5984.

40. Mantey SA, Coy DH, Pradhan TK, Igarashi H, Rizo IM, Shen L, Hou W, Hocart SJ, Jensen RT. Rational design of a peptide agonist that interacts selectively with the orphan receptor, bombesin receptor sub-type 3. J Biol Chem 2001;276:9219-9229.

41. Caggiari L, Zanussi S, Crepaldi C, Bortolin MT, Caffau C, D'Andrea M, De Paoli P. Different rates of CD4^sup +^ and CD8^sup +^ T-cell proliferation in interleukin-2-treated human immunodeficiency virus-positive subjects. Cytometry 2001;46:233-237.

42. Keng PC, Siemann DW. Measurement of proliferation activities in human tumor models: a comparison of flow cytometric methods. Radiat Oncol Investig 1998;6:120-127.

43. Jensen RT, Mrozinski JE, Coy DH. Bombesin receptor antagonists: different classes and cellular basis of action. Recent Results Cancer Res 1993;129:87-113.

44. Lyle RE, Tryka AF, Griffin WS, Taylor BJ. Tryptase immunoreactive mast cell hyperplasia in bronchopulmonary dysplasia. Pediatr Pulmonol 1995;19:336-343.

45. Tokita K, Hocart SJ, Coy DH, Jensen RT. Molecular basis of the selectivity of gastrin-releasing peptide receptor for gastrin-releasing peptide. Mol Pharmacol 2002;61:1435-1443.

46. Moody T, Staley J, Zia F, Coy D, Jensen R. Neuromedin B binds with high affinity, elevates cytosolic calcium and stimulates the growth of small-cell lung cancer cell lines. J Pharmacol Exp Ther 1992;263:311-317.

47. Battey J, Wada E. Two distinct receptor subtypes for mammalian bombesin-like peptides. Trends Neurosci 1991;14:524-528.

48. Chang L, Subramaniam M, Yoder BA, Day BJ, Coalson JJ, Sunday ME, Crapo JD. A catalytic antioxidant attenuates alveolar structural remodeling in bronchopulmonary dysplasia. Am J Respir Crit Care Med 2003;167:57-64.

49. Haley KJ, Patidar K, Zhang F, Emanuel RL, Sunday ME. Tumor necrosis factor induces neuroendocrine differentiation in small cell lung carcinoma cell lines. Am J Physiol 1998;275:L311-L321.

50. Emanuel RL, Torday JS, Mu Q, Asokananthan N, Sikorski KA, Sunday ME. Bombesin-like peptides and receptors in normal fetal baboon lung: roles in lung growth and maturation. Am J Physiol 1999;277:L1003-L1017.

51. King KA, Torday JS, Sunday ME. Bombesin and [Leu^sup 8^]phyllolitorin promote fetal mouse lung branching morphogenesis via a specific receptor-mediated mechanism. Proc Natl A cad Sci USA 1995;92:4357-4361.

52. Nakatani Y. Pulmonary endocrine cells in infancy and childhood. Ped Pathol 1991;11:31-48.

53. Elizegi E, Pino I, Vicent S, Blanco D, Saffiotti U, Montuenga LM. Hyperplasia of alveolar neuroendocrine cells in rat lung carcinogenesis by silica with selective expression of proadrenomedullin-derived peptides and amidating enzymes. Lab Invest 2001;81:1627-L638.

54. Aguayo SM, Miller YE, Waldron JA, Sunday ME, Staton GW, King TE. Brief Report: idiopathic diffuse hyperplasia of pulmonary neuroendocrine cells and airways disease. N Engl J Med 1992;327:1285-1288.

55. Aguayo SM. Pulmonary neuroendocrine cells in tobacco-related lung disorders. Anat Rec 1993;236:122-127.

56. Carroll NG, Mutavdzic S, James AL. Distribution and degranulation of airway mast cells in normal and asthmatic subjects. Eur Respir J 2002;19:879-885.

57. Balzar S, Wenzel SE, Chu HW. Transbronchial biopsy as a tool to evaluate small airways in asthma. Eur Respir J 2002;20:254-259.

58. Johnson DE, Georgieff MK. Pulmonary neuroendocrine cells, their secretory products and their potential roles in health and chronic lung disease in infancy. Am Rev Respir Dis 1989;140:1807-1812.

59. Weber S, Babina G, Henz BM. Human leukemic (HMC-1) and normal skin mast cells express [beta]^sub 2^-integrins: characterization of [beta]^sub 2^-integrins and ICAM-1 on HMC-1 cells. Scand J Immunol 1996;45:471-481.

60. Olsson N, Rak S, Nilsson G. Demonstration of mast cell chemotactic activity in bronchoalveolar lavage fluid collected from asthmatic patients before and during pollen season. Clin Immunol 2000;105:455-461.

61. Temann UA, Geba GP, Rankin JA, Flavell RA. Expression of interleukin 9 in the lungs of transgenic mice causes airway inflammation, mast cell hyperplasia, and bronchial hyperresponsiveness. J Exp Med 1998;188:1307-1320.

62. Galli SJ, Gordon JR, Wershil BK. Mast cell cytokines in allergy and inflammation. Agents Actions 1993;43:209-220.

63. Akers IA, Parsons M, Hill MR, Hollenberg MD, Sanjar S. Laurent GJ, McAnulty RJ. Mast cell tryptase stimulates human lung fibroblast proliferation via protease-activatcd receptor-2. Am J Physiol 2000;278:L193-L201.

64. Groneck P, Speer CP. Pulmonary inflammation in the pathogenesis of bronchopulmonary dysplasia. Pediatr Pulmonol Suppl 1997;16:29-30.

65. Raghavender B, Smith JB. Eosinophil cationic protein in tracheal aspirates of preterm infants with bronchopulmonary dysplasia. J Pediatr 1997;130:944-947.

66. Benya R, Kusui T, Pradhan T, Battey J, Jensen R. Expression and characterization of cloned human bombesin receptors. Mol Pharmacol 1995;47:10-20.

67. Ryan RR, Weber HC, Hou W, Sainz E, Mantey SA, Battey JF, Coy DH, Jensen RT. Ability of various bombesin receptor agonists and antagonists to alter intracellular signaling of the human orphan receptor BRS-3. J Biol Chem 1998;273:13613-13624.

68. Coy DH, Mungan Z, Rossowski WJ, Cheng BL, Lin JT, Mrozinski JE Jr. Jensen RT. Development of a potent bombesin receptor antagonist with prolonged in vivo inhibitory activity on bombesin-stimulated amylase and protein release in the rat. Peptides 1992;13:775-781.

69. Aguayo SM, Kane MA, King TE, Schwarz MI, Grauer L, Miller YE. Increased levels of bombesin-like peptides in the lower respiratory tract of asymptomatic cigarette smokers. J Clin Invest 1989;84:1105-1113.

70. Moody TW, Jensen RT, Garcia L, Leyton J. Nonpeptide neuromedin B receptor antagonists inhibit the proliferation of C6 cells. Eur J Pharmacol 2000;409:133-142.

71. Burghardt B, Wenger C, Barabas K, Racz G, Olah A, Flautner L, Coy DH, Gress TM, Varga G. GRP-receptor-mediated signal transduction, gene expression and DNA synthesis in the human pancreatic adenocarcinoma cell line HPAF. Peptides 2001;22:1119-1128.

72. Nickerson BG, Taussig LM. Family history of asthma in infants with bronchopulmonary dysplasia. Pediatrics 1980;65:1140-1144.

73. Evans M, Palta M, Sadek M, Weinstein MR, Peters ME. Associations between family history of asthma, bronchopulmonary dysplasia, and childhood asthma in very low birth weight children. Am J Epidemiol 1998;148:460-466.

74. Motoyama EK, Fort MD, Klesch KW, Mutich RL, Guthrie RD. Early onset of airway reactivity in premature infants with bronchopulmonary dysplasia. Am Rev Respir Dis 1987;136:50-57.

Meera Subramaniam, Kumiya Sugiyama, David H. Coy, Yanping Kong, York E. Miller, Peter F. Weller, Keiji Wada, Etsuko Wada, and Mary E. Sunday

Department of Pathology and Pulmonology, Children's Hospital Boston; Department of Medicine, Harvard Thorndike Laboratories, Charles A. Dana Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School; and Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts; Department of Medicine, Peptide Research, Tulane University Health Sciences, New Orleans, Louisiana; Department of Medicine, Veterans Affairs Medical Center, University of Colorado, Denver, Colorado; and Institute of Neuroscience, Tokyo, Japan

(Received in original form December 9, 2002; accepted in final form June 4, 2003)

Supported by NIH grants HL52638 (M.E.S.), HL52636 (Animal Core), HL56386 (P.F.W), and AI20241 (P.F.W.).

Correspondence and requests for reprints should be addressed to Mary E. Sunday, M.D., Ph.D., Brigham and Women's Hospital, Department of Pathology, 75 Francis Street, Boston, MA 02115. E-mail: sunday@tch.harvard.edu

This article has an online supplement, which is accessible from this issue's table of contents online at www.atsjournals.org

Copyright American Thoracic Society Sep 1, 2003
Provided by ProQuest Information and Learning Company. All rights Reserved

Return to Bronchopulmonary dysplasia
Home Contact Resources Exchange Links ebay