Targeted expression of heme oxygenase-1 prevents the pulmonary inflammatory and vascular responses to hypoxia

Chronic hypoxia causes pulmonary hypertension with smooth muscle cell proliferation and matrix deposition in the wall of the pulmonary arterioles. We demonstrate here that hypoxia also induces a pronounced inflammation in the lung before the structural changes of the vessel wall. The proinflammatory action of hypoxia is mediated by the induction of distinct cytokines and chemokines and is independent of tumor necrosis factor-α signaling. We have previously proposed a crucial role for heme oxygenase-1 (HO-1) in protecting cardiomyocytes from hypoxic stress, and potent anti-inflammatory properties of HO-1 have been reported in models of tissue injury. We thus established transgenic mice that constitutively express HO-1 in the lung and exposed them to chronic hypoxia. HO-1 transgenic mice were protected from the development of both pulmonary inflammation as well as hypertension and vessel wall hypertrophy induced by hypoxia. Significantly, the hypoxic induction of proinflammatory cytokines and chemokines was suppressed in HO-1 transgenic mice. Our findings suggest an important protective function of enzymatic products of HO-1 activity as inhibitors of hypoxia-induced vasoconstrictive and proinflammatory pathways.

[1]  Adrian M Heilbut,et al.  Complete reversal of fatal pulmonary hypertension in rats by a serine elastase inhibitor , 2000, Nature Medicine.

[2]  A. Choi,et al.  Carbon monoxide has anti-inflammatory effects involving the mitogen-activated protein kinase pathway , 2000, Nature Medicine.

[3]  J Zaky,et al.  Upregulation of heme oxygenase-1 protects genetically fat Zucker rat livers from ischemia/reperfusion injury. , 1999, The Journal of clinical investigation.

[4]  B. Gewertz,et al.  Endothelial permeability and IL-6 production during hypoxia: role of ROS in signal transduction. , 1999, American journal of physiology. Lung cellular and molecular physiology.

[5]  T. Standiford,et al.  Chemokines in lung injury: Thomas A. Neff Lecture. , 1999, Chest.

[6]  L. Kobzik,et al.  Hypoxia induces severe right ventricular dilatation and infarction in heme oxygenase-1 null mice. , 1999, The Journal of clinical investigation.

[7]  A. Choi,et al.  Exogenous administration of heme oxygenase-1 by gene transfer provides protection against hyperoxia-induced lung injury. , 1999, The Journal of clinical investigation.

[8]  M. Jordana,et al.  Cytokines and pulmonary inflammatory and immune diseases. , 1999, Histology and histopathology.

[9]  N. Chandel,et al.  Mitochondrial reactive oxygen species trigger hypoxia-induced transcription. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[10]  R. Colvin,et al.  Expression of heme oxygenase-1 can determine cardiac xenograft survival , 1998, Nature Medicine.

[11]  I. Charo,et al.  Decreased lesion formation in CCR2−/− mice reveals a role for chemokines in the initiation of atherosclerosis , 1998, Nature.

[12]  S. Kourembanas,et al.  Increased vascular endothelial growth factor production in the lungs of rats with hypoxia-induced pulmonary hypertension. , 1998, American journal of respiratory cell and molecular biology.

[13]  K. Matsushima,et al.  Alleviation of monocrotaline-induced pulmonary hypertension by antibodies to monocyte chemotactic and activating factor/monocyte chemoattractant protein-1. , 1998, Laboratory investigation; a journal of technical methods and pathology.

[14]  M. Jordana,et al.  IL-6 is an antiinflammatory cytokine required for controlling local or systemic acute inflammatory responses. , 1998, The Journal of clinical investigation.

[15]  S. Kourembanas,et al.  Carbon Monoxide Controls the Proliferation of Hypoxic Vascular Smooth Muscle Cells* , 1997, The Journal of Biological Chemistry.

[16]  B. Rollins,et al.  Tissue Factor Is Induced by Monocyte Chemoattractant Protein-1 in Human Aortic Smooth Muscle and THP-1 Cells* , 1997, The Journal of Biological Chemistry.

[17]  M. Maines,et al.  Isolation and characterization of a cDNA from the rat brain that encodes hemoprotein heme oxygenase-3. , 1997, European journal of biochemistry.

[18]  P. Jones,et al.  Tenascin-C, proliferation and subendothelial fibronectin in progressive pulmonary vascular disease. , 1997, The American journal of pathology.

[19]  Y. S. Zhou,et al.  Monocytes and tissue factor promote thrombosis in a murine model of oxygen deprivation. , 1997, The Journal of clinical investigation.

[20]  S. Kourembanas,et al.  Mechanisms by which oxygen regulates gene expression and cell-cell interaction in the vasculature. , 1997, Kidney international.

[21]  M. Rabinovitch,et al.  Exogenous leukocyte and endogenous elastases can mediate mitogenic activity in pulmonary artery smooth muscle cells by release of extracellular matrix‐bound basic fibroblast growth factor , 1996, Journal of cellular physiology.

[22]  N. Abraham,et al.  The Biological Significance and Physiological Role of Heme Oxygenase , 1996 .

[23]  S. Kourembanas,et al.  Endothelial cell expression of vasoconstrictors and growth factors is regulated by smooth muscle cell-derived carbon monoxide. , 1995, The Journal of clinical investigation.

[24]  M. Gerritsen,et al.  Transfection of the human heme oxygenase gene into rabbit coronary microvessel endothelial cells: protective effect against heme and hemoglobin toxicity. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[25]  M. Humbert,et al.  Increased interleukin-1 and interleukin-6 serum concentrations in severe primary pulmonary hypertension. , 1995, American journal of respiratory and critical care medicine.

[26]  S. Kourembanas,et al.  Smooth muscle cell-derived carbon monoxide is a regulator of vascular cGMP. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[27]  M. Nauck,et al.  Intracellular interleukin 6 mediates platelet-derived growth factor-induced proliferation of nontransformed cells. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[28]  N. Voelkel,et al.  Interleukin-1 receptor antagonist treatment reduces pulmonary hypertension generated in rats by monocrotaline. , 1994, American journal of respiratory cell and molecular biology.

[29]  T. Yue,et al.  Interleukin-8. A mitogen and chemoattractant for vascular smooth muscle cells. , 1994, Circulation research.

[30]  B. Groves,et al.  Exuberant endothelial cell growth and elements of inflammation are present in plexiform lesions of pulmonary hypertension. , 1994, The American journal of pathology.

[31]  M. Rabinovitch,et al.  Chronic hypoxic pulmonary hypertension in rats and increased elastolytic activity. , 1991, The American journal of physiology.

[32]  S. Potter,et al.  Cis-acting sequences from a human surfactant protein gene confer pulmonary-specific gene expression in transgenic mice. , 1990, Proceedings of the National Academy of Sciences of the United States of America.

[33]  R. Mecham,et al.  Smooth muscle-mediated connective tissue remodeling in pulmonary hypertension. , 1987, Science.

[34]  W. Zapol,et al.  Pulmonary hypertension in severe acute respiratory failure. , 1977, The New England journal of medicine.