Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2009 Oct;297(4):L687-97.
doi: 10.1152/ajplung.90509.2008. Epub 2009 Jul 24.

A paradoxical protective role for the proinflammatory peptide substance P receptor (NK1R) in acute hyperoxic lung injury

Affiliations

A paradoxical protective role for the proinflammatory peptide substance P receptor (NK1R) in acute hyperoxic lung injury

Marwan Dib et al. Am J Physiol Lung Cell Mol Physiol. 2009 Oct.

Abstract

The neuropeptide substance P manifests its biological functions through ligation of a G protein-coupled receptor, the NK1R. Mice with targeted deletion of this receptor reveal a preponderance of proinflammatory properties resulting from ligand activation, demonstrating a neurogenic component to multiple forms of inflammation and injury. We hypothesized that NK1R deficiency would afford a similar protection from inflammation associated with hyperoxia. Counter to our expectations, however, NK1R-/- animals suffered significantly worse lung injury compared with wild-type mice following exposure to 90% oxygen. Median survival was shortened to 84 h for NK1R-/- mice from 120 h for wild-type animals. Infiltration of inflammatory cells into the lungs was significantly increased; NK1R-/- animals also exhibited increased pulmonary edema, hemorrhage, and bronchoalveolar lavage fluid protein levels. TdT-mediated dUTP nick end labeling (TUNEL) staining was significantly elevated in NK1R-/- animals following hyperoxia. Furthermore, induction of metallothionein and Na(+)-K(+)-ATPase was accelerated in NK1R-/- compared with wild-type mice, consistent with increased oxidative injury and edema. In cultured mouse lung epithelial cells in 95% O(2), however, addition of substance P promoted cell death, suggesting the neurogenic component of hyperoxic lung injury is mediated by additional mechanisms in vivo. Release of bioactive constituents including substance P from sensory neurons results from activation of the vanilloid receptor, TRPV1. In mice with targeted deletion of the TRPV1 gene, acute hyperoxic injury is attenuated relative to NK1R-/- animals. Our findings thus reveal a major neurogenic mechanism in acute hyperoxic lung injury and demonstrate concerted actions of sensory neurotransmitters revealing significant protection for NK1R-mediated functions.

PubMed Disclaimer

Figures

Fig. 1.
Fig. 1.
Mortality of neurokinin-1 (NK1) receptor (NK1R) −/− mice is significantly greater than wild-type (WT) animals following exposure to hyperoxia. A: NK1R−/− mice on the C57BL/6 background and wild-type C57BL/6 animals of 8–12 wk were exposed to 90% O2-10% N2, and monitored for mortality. NK1R−/− animals began to die after 72 h, and 100% were dead by 96 h. Initial mortality of wild-type mice was observed at 96 h, with 100% dead by 144 h (different at P < 0.0001; n = 10–16 mice per group). B: posthyperoxia mortality was determined by exposing NK1R−/− and wild-type mice to 90% O2 for 72 h followed by return to room air. NK1R−/− mice began to die within 24 h following 72-h hyperoxia, and mortality reached 70% by 96 h compared with none for the wild-type animals (different at P < 0.005; n = 10 mice per group).
Fig. 2.
Fig. 2.
Microscopic evaluation reveals increased tissue damage in NK1R−/− mouse lungs following hyperoxia relative to wild-type animals. Photomicrographs of NK1R−/− and wild-type mouse lungs exposed to 90% O2 for 72 h are shown. The epithelial injury (*), inflammatory cell infiltrate (▴), increased alveolar thickness (↑), and entrapped red blood cells (▵) characteristic of hyperoxic injury are all significantly more pronounced in NK1R−/− mice compared with wild-type animals. Representative of 6–8 mice per group. Hematoxylin and eosin (H&E) stain, magnification ×400.
Fig. 3.
Fig. 3.
NK1R−/− mice develop more airway edema and bronchoalveolar lavage (BAL) fluid protein than wild-type animals following hyperoxia. A: lung wet/dry weights (*P < 0.0001 for hyperoxia vs. room air; **P < 0.001 for NK1R−/− vs. wild-type mice exposed to hyperoxia; n = 10 mice per group). B: increased microvascular permeability assessed by extravasation of Evans blue dye (*P < 0.05 for hyperoxia vs. room air, wild-type mice; **P < 0.0003 for NK1R−/− vs. wild-type animals after hyperoxia; n = 5–7 mice per group). C: BAL fluid protein concentration (*P < 0.0001 for hyperoxia vs. room air; **P < 0.001 for hyperoxia-exposed NK1R−/− mice vs. wild-type; n = 6–12 mice per group).
Fig. 4.
Fig. 4.
Inflammatory cell influx and hemoglobin content of NK1R−/− and wild-type mouse lungs following hyperoxia. A: the content of neutrophils in BAL fluid is elevated for wild-type but not NK1R−/− mice after exposure to hyperoxia [*P < 0.002 for wild-type mice, hyperoxia vs. room air; **no significant difference (NS) for NK1R−/− mice, hyperoxia vs. room air; n = 11–12 mice per group]. B: lung content of neutrophils determined from the tissue content of MPO (*P < 0.01 for wild-type mice, hyperoxia vs. room air; n = 6–12 mice per group; **P < 0.0001 for hyperoxia-exposed NK1R−/− vs. wild-type mice; n = 11–12 mice per group). C: lung content of hemoglobin (*P < 0.0001 for hyperoxia vs. room air; n = 6–8 mice per group; **P < 0.0005 for NK1R−/− vs. wild-type mice exposed to hyperoxia; n = 6–12 mice per group).
Fig. 5.
Fig. 5.
Hyperoxia-mediated induction of metallothionein and Na+-K+-ATPase is accelerated in NK1R−/− mice compared with wild-type. A: densitometric quantitation of Western blots for metallothionein in the lungs of wild-type and NK1R−/− mice as a function of exposure to 90% O2. Enzyme levels were increased relative to room air-exposed mice as early as 24 h after initiation of hyperoxic conditions for NK1R−/− animals (*P < 0.0001 for NK1R−/− vs. wild-type mice at 24 h; n = 6–8 mice per group). After 48- and 72-h hyperoxia exposure, both NK1R−/− and wild-type mice exhibited elevated metallothionein levels relative to room air, but the levels for NK1R−/− and wild-type animals was not distinguishable (**P < 0.0005 at 24 h, ***P < 0.001 at 72 h, hyperoxia vs. room air, NS for NK1R−/− vs. wild-type mice in hyperoxia; n = 6–8 mice per group). Data are expressed as the percent ± SE of room air-exposed animals. B: representative Western blot of metallothionein extracted from mouse lungs as a function of exposure to hyperoxia. Lung homogenate protein, 25 μg per lane, was separated by SDS-PAGE and Western blotted as described in experimental procedures. The enzyme appears as the monomer and β-ME-resistant oligomers. C: Na+-K+-ATPase levels in lung homogenates were determined by Western blotting and quantitated by densitometry. Values represent the mean percent ± SE relative to room air-exposed mice (*P < 0.0001 for NK1R−/− vs. wild-type mice at 48-h hyperoxia; **P < 0.05 for wild-type mice at 72-h vs. 48-h hyperoxia; n = 5–8 mice per group). D: representative Western blot of Na+-K+-ATPase extracted from mouse lungs as a function of exposure to hyperoxia. Lung homogenate protein, 25 μg per lane, was separated by SDS-PAGE and Western blotted as described in experimental procedures.
Fig. 6.
Fig. 6.
Cytokine levels are decreased for NK1R−/− mice compared with wild-type animals following 72-h hyperoxia. ELISA assays of TNFα, IFN-γ, and IL-1β in homogenates of NK1R−/− and wild-type mouse lungs housed in room air or 90% O2 for 72 h (TNFα, *P < 0.05 for NK1R−/− vs. wild-type mice exposed to hyperoxia; IFN-γ, *P < 0.0001 for both strains, hyperoxia vs. room air, **P < 0.005 for NK1R−/− vs. wild-type mice exposed to hyperoxia; IL-1β, *P < 0.0001 for both strains, room air vs. hyperoxia, **P < 0.0001 for NK1R−/− vs. wild-type mice exposed to hyperoxia; n = 8–15 mice per group).
Fig. 7.
Fig. 7.
NK1R−/− mice exhibit greater evidence of apoptosis compared with wild-type animals after exposure to hyperoxia. A: representative TdT-mediated dUTP nick end labeling (TUNEL) staining of sections of lung tissues from wild-type and NK1R−/− mice housed in room air or 90% O2 for 72 h. Cobalt-enhanced peroxidase staining (black) indicates apoptotic cells (representative of 3 animals per group). Sections were counterstained with nuclear fast red, magnification ×400. B: TUNEL-positive nuclei were enumerated from 20 randomly chosen fields, and 2,000 nuclei per lung were counted. Data are presented as the percent positive cells (*P < 0.05 for wild-type mice, hyperoxia vs. room air; **P < 0.0001 for NK1R−/− vs. wild-type following 72-h hyperoxia; n = 3 mice per group). C: enzymatic activity of caspase-3 in lung tissues from mice housed in room air or 90% O2 for 72 h. Equal amounts of protein were assessed colorimetrically for caspase-3 activity by reacting with N-acetyl-Asp-Glu-Val-Asp-p-nitroanilide (Ac-DEVD-pNA) as described in experimental procedures. Data are expressed as the means ± SE picomoles per minute per milligram protein (*P < 0.0001 for both strains, hyperoxia vs. room air; **NS between hyperoxia-exposed NK1R−/− and wild-type mice; n = 4–14 mice per group).
Fig. 8.
Fig. 8.
Cytotoxicity of substance P in NK1R-transfected mouse lung epithelial cells cultured in 95% O2. Murine lung epithelial MLE-12 cells stably transfected with the human NK1R were grown in room air with 5% CO2 or 95% O2-5% CO2 for 24 h in the presence or absence of 1 μM substance P (SP). Cells were harvested, stained with Alexa Fluor 488 annexin V and propidium iodide, and analyzed by flow cytometry. The percent of gated cells in each quadrant is indicated. Representative of 3 independent experiments.
Fig. 9.
Fig. 9.
Survival of transient receptor potential vanilloid type 1 (TRPV1) −/− mice in hyperoxia is increased relative to NK1R−/− animals and indistinguishable from wild-type mice. Mice of the genotypes indicated were placed in 90% O2-10% N2 and monitored for mortality. Survival of TRPV1 mice was significantly increased compared with NK1R−/− animals (P < 0.0001; n = 12 mice per group) but not different from wild-type mice (P = NS; n = 12 mice per group).

Similar articles

Cited by

References

    1. Asehnoune K, Strassheim D, Mitra S, Kim JY, Abraham E. Involvement of reactive oxygen species in Toll-like receptor 4-dependent activation of NF-kappa B. J Immunol 172: 2522–2529, 2004 - PubMed
    1. Barazzone C, Horowitz S, Donati YR, Rodriguez I, Piguet PF. Oxygen toxicity in mouse lung: pathways to cell death. Am J Respir Cell Mol Biol 19: 573–581, 1998 - PubMed
    1. Barbato JC, Catanescu O, Murray K, DiBello PM, Jacobsen DW. Targeting of metallothionein by l-homocysteine: a novel mechanism for disruption of zinc and redox homeostasis. Arterioscler Thromb 27: 49–54, 2007 - PMC - PubMed
    1. Barker GF, Manzo ND, Cotich KL, Shone RK, Waxman AB. DNA damage induced by hyperoxia: quantitation and correlation with lung injury. Am J Respir Cell Mol Biol 35: 277–288, 2006 - PMC - PubMed
    1. Bill A, Stjernschantz J, Mandahl A, Brodin E, Nilsson G. Substance P: release on trigeminal nerve stimulation, effects in the eye. Acta Physiol Scand 106: 371–373, 1979 - PubMed

Publication types

MeSH terms

LinkOut - more resources