51
|
Jernigan NL, Resta TC. Calcium Homeostasis and Sensitization in Pulmonary Arterial Smooth Muscle. Microcirculation 2014; 21:259-71. [DOI: 10.1111/micc.12096] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 09/25/2013] [Indexed: 01/10/2023]
Affiliation(s)
- Nikki L. Jernigan
- Vascular Physiology Group; Department of Cell Biology and Physiology; University of New Mexico Health Sciences Center; Albuquerque New Mexico USA
| | - Thomas C. Resta
- Vascular Physiology Group; Department of Cell Biology and Physiology; University of New Mexico Health Sciences Center; Albuquerque New Mexico USA
| |
Collapse
|
52
|
Peng W, Zhou Q, Ao X, Tang R, Xiao Z. Inhibition of Rho-kinase alleviates peritoneal fibrosis and angiogenesis in a rat model of peritoneal dialysis. Ren Fail 2014; 35:958-66. [PMID: 23859538 DOI: 10.3109/0886022x.2013.808565] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND/AIMS The present study investigated whether Rho-kinase inhibition had a therapeutic role on the pathogenesis of peritoneal fibrosis and angiogenesis. METHODS A rat model of peritoneal dialysis was induced by a daily intraperitoneal infusion of 4.25% Dianeal. Those rats were treated with Rho-kinase inhibitor, fasudil. Immunofluorescence, Western blot and RT-PCR were used to detect the expression of TGF-β1, Collagen I, αSMA and VEGF in each group. Microvessel density (MVD) was measured by immunohistochemistry. Rho-kinase activity was determined by western immunoblotting. RESULTS Rho-kinase was activated in the peritoneum of the PD group, which was inhibited by fasudil. Compared with PD group, the mRNA and protein expressions of TGF-β1, αSMA and Collagen I were significantly downregulated in fasudil treatment groups in a dose-dependent manner, and the expression of VEGF and peritoneal MVD was also significantly downregulated in fasudil treatment groups in a dose-dependent manner. CONCLUSION The Rho-kinase was activated in the peritoneum of the peritoneal dialysis rats, and the inhibition of Rho-kinase by fasudil can remarkably decrease peritoneal fibrosis and angiogenesis.
Collapse
Affiliation(s)
- Weisheng Peng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | | | | | | | | |
Collapse
|
53
|
Ball MK, Waypa GB, Mungai PT, Nielsen JM, Czech L, Dudley VJ, Beussink L, Dettman RW, Berkelhamer SK, Steinhorn RH, Shah SJ, Schumacker PT. Regulation of hypoxia-induced pulmonary hypertension by vascular smooth muscle hypoxia-inducible factor-1α. Am J Respir Crit Care Med 2014. [PMID: 24251580 DOI: 10.1164/rccm.201302-03020c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
RATIONALE Chronic hypoxia induces pulmonary vascular remodeling, pulmonary hypertension, and right ventricular hypertrophy. At present, little is known about mechanisms driving these responses. Hypoxia-inducible factor-1α (HIF-1α) is a master regulator of transcription in hypoxic cells, up-regulating genes involved in energy metabolism, proliferation, and extracellular matrix reorganization. Systemic loss of a single HIF-1α allele has been shown to attenuate hypoxic pulmonary hypertension, but the cells contributing to this response have not been identified. OBJECTIVES We sought to determine the contribution of HIF-1α in smooth muscle on pulmonary vascular and right heart responses to chronic hypoxia. METHODS We used mice with homozygous conditional deletion of HIF-1α combined with tamoxifen-inducible smooth muscle-specific Cre recombinase expression. Mice received either tamoxifen or vehicle followed by exposure to either normoxia or chronic hypoxia (10% O2) for 30 days before measurement of cardiopulmonary responses. MEASUREMENTS AND MAIN RESULTS Tamoxifen-induced smooth muscle-specific deletion of HIF-1α attenuated pulmonary vascular remodeling and pulmonary hypertension in chronic hypoxia. However, right ventricular hypertrophy was unchanged despite attenuated pulmonary pressures. CONCLUSIONS These results indicate that HIF-1α in smooth muscle contributes to pulmonary vascular remodeling and pulmonary hypertension in chronic hypoxia. However, loss of HIF-1 function in smooth muscle does not affect hypoxic cardiac remodeling, suggesting that the cardiac hypertrophy response is not directly coupled to the increase in pulmonary artery pressure.
Collapse
|
54
|
Ball MK, Waypa GB, Mungai PT, Nielsen JM, Czech L, Dudley VJ, Beussink L, Dettman RW, Berkelhamer SK, Steinhorn RH, Shah SJ, Schumacker PT. Regulation of hypoxia-induced pulmonary hypertension by vascular smooth muscle hypoxia-inducible factor-1α. Am J Respir Crit Care Med 2014; 189:314-24. [PMID: 24251580 DOI: 10.1164/rccm.201302-0302oc] [Citation(s) in RCA: 208] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
RATIONALE Chronic hypoxia induces pulmonary vascular remodeling, pulmonary hypertension, and right ventricular hypertrophy. At present, little is known about mechanisms driving these responses. Hypoxia-inducible factor-1α (HIF-1α) is a master regulator of transcription in hypoxic cells, up-regulating genes involved in energy metabolism, proliferation, and extracellular matrix reorganization. Systemic loss of a single HIF-1α allele has been shown to attenuate hypoxic pulmonary hypertension, but the cells contributing to this response have not been identified. OBJECTIVES We sought to determine the contribution of HIF-1α in smooth muscle on pulmonary vascular and right heart responses to chronic hypoxia. METHODS We used mice with homozygous conditional deletion of HIF-1α combined with tamoxifen-inducible smooth muscle-specific Cre recombinase expression. Mice received either tamoxifen or vehicle followed by exposure to either normoxia or chronic hypoxia (10% O2) for 30 days before measurement of cardiopulmonary responses. MEASUREMENTS AND MAIN RESULTS Tamoxifen-induced smooth muscle-specific deletion of HIF-1α attenuated pulmonary vascular remodeling and pulmonary hypertension in chronic hypoxia. However, right ventricular hypertrophy was unchanged despite attenuated pulmonary pressures. CONCLUSIONS These results indicate that HIF-1α in smooth muscle contributes to pulmonary vascular remodeling and pulmonary hypertension in chronic hypoxia. However, loss of HIF-1 function in smooth muscle does not affect hypoxic cardiac remodeling, suggesting that the cardiac hypertrophy response is not directly coupled to the increase in pulmonary artery pressure.
Collapse
|
55
|
Gien J, Tseng N, Seedorf G, Roe G, Abman SH. Peroxisome proliferator activated receptor-γ-Rho-kinase interactions contribute to vascular remodeling after chronic intrauterine pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2013; 306:L299-308. [PMID: 24375792 DOI: 10.1152/ajplung.00271.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Peroxisome proliferator-activated receptor-γ (PPARγ) and Rho-kinase (ROCK) regulate smooth muscle cell (SMC) proliferation and contribute to vascular remodeling in adult pulmonary hypertension. Whether these pathways interact to contribute to the development of vascular remodeling in persistent pulmonary hypertension of the newborn (PPHN) remains unknown. We hypothesized that ROCK-PPARγ interactions increase SMC proliferation resulting in vascular remodeling in experimental PPHN. Pulmonary artery SMCs (PASMCs) were harvested from fetal sheep after partial ligation of the ductus arteriosus in utero (PPHN) and controls. Cell counts were performed daily for 5 days with or without PPARγ agonists and ROCK inhibition. PPARγ and ROCK protein expression/activity were measured by Western blot in normal and PPHN PASMCs. We assessed PPARγ-ROCK interactions by studying the effect of ROCK activation on PPARγ activity and PPARγ inhibition (siRNA) on ROCK activity and PASMC proliferation. At baseline, PPHN PASMC cell number was increased by 38% above controls on day 5. ROCK protein expression/activity were increased by 25 and 34% and PPARγ protein/activity decreased by 40 and 50% in PPHN PASMC. ROCK inhibition and PPARγ activation restored PPHN PASMC growth to normal values. ROCK inhibition increased PPARγ activity by 50% in PPHN PASMC, restoring PPARγ activity to normal. In normal PASMCs, ROCK activation decreased PPARγ activity and PPARγ inhibition increased ROCK activity and cell proliferation, resulting in a PPHN hyperproliferative PASMC phenotype. PPARγ-ROCK interactions regulate SMC proliferation and contribute to increased PPHN PASMC proliferation and vascular remodeling in PPHN. Restoring normal PPARγ-ROCK signaling may prevent vascular remodeling and improve outcomes in PPHN.
Collapse
Affiliation(s)
- Jason Gien
- Perinatal Research Facility, 13243 E. 23rd Ave., Mail Stop F441, Aurora, CO 80045.
| | | | | | | | | |
Collapse
|
56
|
Swain SD, Siemsen DW, Pullen RR, Han S. CD4+ T cells and IFN-γ are required for the development of Pneumocystis-associated pulmonary hypertension. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 184:483-93. [PMID: 24361497 DOI: 10.1016/j.ajpath.2013.10.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 09/11/2013] [Accepted: 10/23/2013] [Indexed: 12/24/2022]
Abstract
Pulmonary hypertension (PH) is a disease of diverse etiology. Although primary PH can develop in the absence of prior disease, PH more commonly develops in conjunction with other pulmonary pathologies. We previously reported a mouse model in which PH occurs as a sequela of Pneumocystis infection in the context of transient CD4 depletion. Here, we report that instead of the expected Th2 pathways, the Th1 cytokine IFN-γ is essential for the development of PH, as wild-type mice developed PH but IFN-γ knockout mice did not. Because gene expression analysis showed few strain differences that were not immune-function related, we focused on those responses as potential pathologic mechanisms. In addition to dependence on IFN-γ, we found that when CD4 cells were continuously depleted, but infection was limited by antibiotic treatment, PH did not occur, confirming that CD4 T cells are required for PH development. Also, although CD8 T-cells are implicated in the pathology of Pneumocystis pneumonia, they did not have a role in the onset of PH. Finally, we found differences in immune cell phenotypes that correlated with PH, including elevated CD204 expression in lung CD11c(+) cells, but their role remains unclear. Overall, we demonstrate that a transient, localized, immune response requiring IFN-γ and CD4-T cells can disrupt pulmonary vascular function and promote lingering PH.
Collapse
Affiliation(s)
- Steve D Swain
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana.
| | - Dan W Siemsen
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana
| | - Rebecca R Pullen
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana
| | - Soo Han
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana
| |
Collapse
|
57
|
McLoughlin P, Keane MP. Physiological and pathological angiogenesis in the adult pulmonary circulation. Compr Physiol 2013; 1:1473-508. [PMID: 23733650 DOI: 10.1002/cphy.c100034] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Angiogenesis occurs during growth and physiological adaptation in many systemic organs, for example, exercise-induced skeletal and cardiac muscle hypertrophy, ovulation, and tissue repair. Disordered angiogenesis contributes to chronic inflammatory disease processes and to tumor growth and metastasis. Although it was previously thought that the adult pulmonary circulation was incapable of supporting new vessel growth, over that past 10 years new data have shown that angiogenesis within this circulation occurs both during physiological adaptive processes and as part of the pathogenic mechanisms of lung diseases. Here we review the expression of vascular growth factors in the adult lung, their essential role in pulmonary vascular homeostasis and the changes in their expression that occur in response to physiological challenges and in disease. We consider the evidence for adaptive neovascularization in the pulmonary circulation in response to alveolar hypoxia and during lung growth following pneumonectomy in the adult lung. In addition, we review the role of disordered angiogenesis in specific lung diseases including idiopathic pulmonary fibrosis, acute adult distress syndrome and both primary and metastatic tumors of the lung. Finally, we examine recent experimental data showing that therapeutic enhancement of pulmonary angiogenesis has the potential to treat lung diseases characterized by vessel loss.
Collapse
Affiliation(s)
- Paul McLoughlin
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, and St. Vincent's University Hospital, Dublin, Ireland.
| | | |
Collapse
|
58
|
Li L, Howell K, Sands M, Banahan M, Frohlich S, Rowan SC, Neary R, Ryan D, McLoughlin P. The α and Δ isoforms of CREB1 are required to maintain normal pulmonary vascular resistance. PLoS One 2013; 8:e80637. [PMID: 24349008 PMCID: PMC3857174 DOI: 10.1371/journal.pone.0080637] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 10/05/2013] [Indexed: 01/15/2023] Open
Abstract
Chronic hypoxia causes pulmonary hypertension associated with structural alterations in pulmonary vessels and sustained vasoconstriction. The transcriptional mechanisms responsible for these distinctive changes are unclear. We have previously reported that CREB1 is activated in the lung in response to alveolar hypoxia but not in other organs. To directly investigate the role of α and Δ isoforms of CREB1 in the regulation of pulmonary vascular resistance we examined the responses of mice in which these isoforms of CREB1 had been inactivated by gene mutation, leaving only the β isoform intact (CREB(αΔ) mice). Here we report that expression of CREB regulated genes was altered in the lungs of CREB(αΔ) mice. CREB(αΔ) mice had greater pulmonary vascular resistance than wild types, both basally in normoxia and following exposure to hypoxic conditions for three weeks. There was no difference in rho kinase mediated vasoconstriction between CREB(αΔ) and wild type mice. Stereological analysis of pulmonary vascular structure showed characteristic wall thickening and lumen reduction in hypoxic wild-type mice, with similar changes observed in CREB(αΔ). CREB(αΔ) mice had larger lungs with reduced epithelial surface density suggesting increased pulmonary compliance. These findings show that α and Δ isoforms of CREB1 regulate homeostatic gene expression in the lung and that normal activity of these isoforms is essential to maintain low pulmonary vascular resistance in both normoxic and hypoxic conditions and to maintain the normal alveolar structure. Interventions that enhance the actions of α and Δ isoforms of CREB1 warrant further investigation in hypoxic lung diseases.
Collapse
Affiliation(s)
- Lili Li
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
| | - Katherine Howell
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
| | - Michelle Sands
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
| | - Mark Banahan
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
| | - Stephen Frohlich
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
- Department of Anaesthesia and Critical Care, St Vincent's University Hospital, Dublin, Ireland
| | - Simon C. Rowan
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
| | - Roisín Neary
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
| | - Donal Ryan
- Department of Anaesthesia and Critical Care, St Vincent's University Hospital, Dublin, Ireland
| | - Paul McLoughlin
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, Dublin, Ireland
| |
Collapse
|
59
|
Garat CV, Crossno JT, Sullivan TM, Reusch JEB, Klemm DJ. Inhibition of phosphatidylinositol 3-kinase/Akt signaling attenuates hypoxia-induced pulmonary artery remodeling and suppresses CREB depletion in arterial smooth muscle cells. J Cardiovasc Pharmacol 2013; 62:539-48. [PMID: 24084215 PMCID: PMC4143163 DOI: 10.1097/fjc.0000000000000014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Hypoxia-induced pulmonary hypertension is characterized by progressive remodeling of the pulmonary artery (PA) system and loss of the transcription factor, cAMP response element binding protein (CREB) in PA smooth muscle cells (SMCs). Previous in vitro studies suggested that platelet-derived growth factor, a mitogen produced in the hypoxic arterial wall, elicits loss of CREB in medial SMCs via the PI3K/Akt pathway. These events trigger switching of SMCs from a quiescent, contractile phenotype to a proliferative, migratory, dedifferentiated, and synthetic phenotype, which contributes to PA thickening. Here, we investigated whether inhibition of PI3K or Akt could attenuate arterial remodeling in the lung and prevent CREB loss in PA medial SMCs in rats subjected to chronic hypoxia. Inhibition of either enzyme-blunted hypoxia-induced PA remodeling and SMC CREB depletion and diminished SMC proliferation and collagen deposition. Inhibition of Akt, but not PI3K, suppressed muscularization of distal arterioles and blunted right ventricular hypertrophy. Interestingly, mean PA pressure was elevated equally by hypoxia in untreated and inhibitor-treated groups but was normalized acutely by the Rho kinase inhibitor, Fasudil. We conclude that PI3K and Akt inhibitors can attenuate hypoxia-induced PA remodeling and SMC CREB depletion but fail to block the development of pulmonary hypertension because of their inability to repress Rho kinase-mediated vasoconstriction.
Collapse
MESH Headings
- Animals
- Arterioles/drug effects
- Arterioles/metabolism
- Arterioles/pathology
- Cell Proliferation/drug effects
- Cyclic AMP Response Element-Binding Protein/agonists
- Cyclic AMP Response Element-Binding Protein/metabolism
- Enzyme Inhibitors/therapeutic use
- Extracellular Matrix/drug effects
- Extracellular Matrix/metabolism
- Extracellular Matrix/pathology
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/prevention & control
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/prevention & control
- Hypoxia/physiopathology
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Phosphatidylinositol 3-Kinase/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Protein Stability/drug effects
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/metabolism
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Circulation/drug effects
- Rats
- Rats, Inbred WKY
- Signal Transduction/drug effects
- Vasodilator Agents/pharmacology
- Vasodilator Agents/therapeutic use
- rho-Associated Kinases/antagonists & inhibitors
- rho-Associated Kinases/metabolism
Collapse
Affiliation(s)
- Chrystelle V. Garat
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Divisions of Pulmonary Science and Critical Care Medicine, University of Colorado Anschutz Medical campus, Aurora, CO
| | - Joseph T. Crossno
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Divisions of Pulmonary Science and Critical Care Medicine, University of Colorado Anschutz Medical campus, Aurora, CO
| | - Timothy M. Sullivan
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jane E. B. Reusch
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Divisions of Endocrinology, University of Colorado Anschutz Medical campus, Aurora, CO
- Research and Endocrine Services, Veterans Affairs Medical Center, Denver, CO
| | - Dwight J. Klemm
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Divisions of Pulmonary Science and Critical Care Medicine, University of Colorado Anschutz Medical campus, Aurora, CO
| |
Collapse
|
60
|
Peng C, Gu P, Zhou J, Huang J, Wang W. Inhibition of rho-kinase by fasudil suppresses formation and progression of experimental abdominal aortic aneurysms. PLoS One 2013; 8:e80145. [PMID: 24244631 PMCID: PMC3828185 DOI: 10.1371/journal.pone.0080145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 10/10/2013] [Indexed: 11/18/2022] Open
Abstract
Objective Accumulating evidence suggests that inflammatory cell infiltration is crucial pathogenesis during the initiation and progression of abdominal aortic aneurysm (AAA). Given Rho-kinase (ROCK), an important kinase control the actin cytoskeleton, regulates the inflammatory cell infiltration, thus, we investigate the possibility and mechanism of preventing experimental AAA progression via targeting ROCK in mice porcine pancreatic elastase (PPE) model. Methods and Results AAA was created in 10-week-old male C57BL/6 mice by transient intraluminal porcine pancreatic elastase infusion into the infrarenal aorta. The mRNA level of RhoA, RhoC, ROCK1 and ROCK2 were elevated in aneurismal aorta. Next, PPE infusion mice were orally administrated with vehicle or ROCK inhibitor (Fasudil at dose of 200 mg/kg/day) during the period of day 1 prior to PPE infusion to day 14 after PPE infusion. PPE infusion mice treated with Fasudil produced significantly smaller aneurysms as compare to PPE infusion mice treated with vehicle. AAAs developed in all vehicle-treated groups within 14 days, whereas AAAs developed in six mice (66%, 6/9) treated with Fasudil within 14 days. Furthermore, our semi-quantitative histological analysis revealed that blood vessels and macrophages were significantly reduced in Fasudil treated mice during the AAA progression. Finally, when mice with existing AAAs were treated with Fasudil, the enlargement was nearly completely suppressed. Conclusion Fasudil inhibits experimental AAA progression and stabilize existing aneurysms, through mechanisms likely related to impaired mural macrophage infiltration and angiogenesis. These findings suggest that ROCK inhibitor may hold substantial translational value for AAA diseases.
Collapse
Affiliation(s)
- Chen Peng
- Department of Vascular Surgery,Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Peng Gu
- Department of Vascular Surgery,Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Zhou
- Department of Stomatolog, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianhua Huang
- Department of Vascular Surgery,Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Wang
- Department of Vascular Surgery,Xiangya Hospital, Central South University, Changsha, Hunan, China
- * E-mail:
| |
Collapse
|
61
|
Bei Y, Duong-Quy S, Hua-Huy T, Dao P, Le-Dong NN, Dinh-Xuan AT. Activation of RhoA/Rho-kinase pathway accounts for pulmonary endothelial dysfunction in patients with chronic obstructive pulmonary disease. Physiol Rep 2013; 1:e00105. [PMID: 24303177 PMCID: PMC3841041 DOI: 10.1002/phy2.105] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 09/03/2013] [Accepted: 09/04/2013] [Indexed: 12/04/2022] Open
Abstract
Recent evidence suggests that activation of RhoA/Rho-kinase accounts for systemic and pulmonary endothelial dysfunction in smokers with normal lung function. However, its role in patients with chronic obstructive pulmonary disease (COPD) has not yet been investigated. The aim of this study was to evaluate the regulation of RhoA/Rho-kinase pathway and pulmonary endothelial dysfunction in patients with COPD. Pulmonary arteries were obtained from nonsmokers (control subjects) and patients with nonhypoxemic and hypoxemic COPD (n = 6–7/group). Endothelium-dependent and -independent relaxations were evaluated by acetylcholine and sodium nitroprusside, respectively. Gene and protein expressions of endothelial nitric oxide synthase (eNOS) were measured by RT-PCR, Western blot, and immunohistochemistry. Nitrate, cGMP, and endothelin-1 (ET-1) concentrations, as well as Rho-kinase activity were measured by ELISA. Protein expressions of total RhoA and GTP-RhoA were measured by Western blot and pull-down assay, respectively. Endothelium-dependent relaxation, and nitrate and cGMP levels were significantly reduced in pulmonary arteries of COPD patients as compared with control subjects. Conversely, activity of RhoA/Rho-kinase was increased in pulmonary arteries of COPD patients as compared with control subjects. In patients with COPD, pulmonary endothelial dysfunction was related to the downregulation of eNOS activity and upregulation of RhoA/Rho-kinase activity.
Collapse
Affiliation(s)
- Yihua Bei
- Medical School, Assistance Publique Hôpitaux de Paris, Service de Physiologie, Paris Descartes University EA 2511, Hôpital Cochin, 27 Rue du faubourg Saint-Jacques, 75014, Paris, France ; Clinical and Translational Research Center, Tongji University School of Medicine and Shanghai East Hospital 150 Jimo Road, Shanghai, 200120, China
| | | | | | | | | | | |
Collapse
|
62
|
Vanderpool RR, El-Bizri N, Rabinovitch M, Chesler NC. Patchy deletion of Bmpr1a potentiates proximal pulmonary artery remodeling in mice exposed to chronic hypoxia. Biomech Model Mechanobiol 2013; 12:33-42. [PMID: 22314711 DOI: 10.1007/s10237-012-0379-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 01/23/2012] [Indexed: 11/28/2022]
Abstract
Reduced vascular expression of bone morphogenetic protein type IA receptor (Bmpr1a) has been found in patients with pulmonary arterial hypertension. Our previous studies in mice with patchy deletion of Bmpr1a in vascular smooth muscle cells and cardiac myocytes showed decreased distal vascular remodeling despite a similar severity of hypoxic pulmonary hypertension (HPH). We speculate increased stiffness from ectopic deposition of collagen in proximal pulmonary arteries might account for HPH. Pulsatile pressure-flow relationships were measured in isolated, ventilated, perfused lungs of SM22α;TRE-Cre;R26R;Bmpr1a(flox/flox) (KO) mice and wild-type littermates, following 21 days (hypoxia) and 0 days (control) of chronic hypoxia. Pulmonary vascular impedance, which yields insight into proximal and distal arterial remodeling, was calculated. Reduced Bmpr1a expression had no effect on input impedance Z(0) (P = 0.52) or characteristic impedance Z(C) (P = 0.18) under control conditions; it also had no effect on the decrease in Z(0) via acute rho kinase inhibition. However, following chronic hypoxia, reduced Bmpr1a expression increased Z(C) (P < 0.001) without affecting Z(0) (P = 0.72). These results demonstrate that Bmpr1a deficiency does not significantly alter the hemodynamic function of the distal vasculature or its response to chronic hypoxia but larger, more proximal arteries are affected. In particular, reduced Bmpr1a expression likely decreased dilatation and increased stiffening in response to hypoxia, probably by collagen accumulation. Increased PA stiffness can have a significant impact on right ventricular function. This study illustrates for the first time how proximal pulmonary artery changes in the absence of distal pulmonary artery changes contribute to pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Rebecca R Vanderpool
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
63
|
Nielsen G, Wandall-Frostholm C, Sadda V, Oliván-Viguera A, Lloyd EE, Bryan RM, Simonsen U, Köhler R. Alterations of N-3 polyunsaturated fatty acid-activated K2P channels in hypoxia-induced pulmonary hypertension. Basic Clin Pharmacol Toxicol 2013; 113:250-8. [PMID: 23724868 DOI: 10.1111/bcpt.12092] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 05/24/2013] [Indexed: 01/06/2023]
Abstract
Polyunsaturated fatty acid (PUFA)-activated two-pore domain potassium channels (K2P ) have been proposed to be expressed in the pulmonary vasculature. However, their physiological or pathophysiological roles are poorly defined. Here, we tested the hypothesis that PUFA-activated K2P are involved in pulmonary vasorelaxation and that alterations of channel expression are pathophysiologically linked to pulmonary hypertension. Expression of PUFA-activated K2P in the murine lung was investigated by quantitative reverse-transcription polymerase chain reaction (qRT-PCR), immunohistochemistry (IHC), by patch clamp (PC) and myography. K2P -gene expression was examined in chronic hypoxic mice. qRT-PCR showed that the K2P 2.1 and K2P 6.1 were the predominantly expressed K2P in the murine lung. IHC revealed protein expression of K2P 2.1 and K2P 6.1 in the endothelium of pulmonary arteries and of K2P 6.1 in bronchial epithelium. PC showed pimozide-sensitive K2P -like K(+) -current activated by docosahexaenoic acid (DHA) in freshly isolated endothelial cells as well as DHA-induced membrane hyperpolarization. Myography on pulmonary arteries showed that DHA induced concentration-dependent instantaneous relaxations that were resistant to endothelial removal and inhibition of NO and prostacyclin synthesis and to a cocktail of blockers of calcium-activated K(+) channels but were abolished by high extracellular (30 mM) K(+) -concentration. Gene expression and protein of K2P 2.1 were not altered in chronic hypoxic mice, while K2P 6.1 was up-regulated by fourfold. In conclusion, the PUFA-activated K2P 2.1 and K2P 6.1 are expressed in murine lung and functional K2P -like channels contribute to endothelium hyperpolarization and pulmonary artery relaxation. The increased K2P 6.1-gene expression may represent a novel counter-regulatory mechanism in pulmonary hypertension and suggest that arterial K2P 2.1 and K2P 6.1 could be novel therapeutic targets.
Collapse
Affiliation(s)
- Gorm Nielsen
- Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
64
|
Cabral JE, Belik J. Persistent pulmonary hypertension of the newborn: Recent advances in pathophysiology and treatment. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2013. [DOI: 10.1016/j.jpedp.2012.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
65
|
Cabral JEB, Belik J. Persistent pulmonary hypertension of the newborn: recent advances in pathophysiology and treatment. J Pediatr (Rio J) 2013; 89:226-42. [PMID: 23684454 DOI: 10.1016/j.jped.2012.11.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 11/08/2012] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Although recognized for decades, little is known about the etiology, physiopathology, and prevention of persistent pulmonary hypertension of the newborn (PPHN), and its treatment remains a major challenge for neonatologists. In this review, the clinical features and physiopathology of the syndrome will be addressed, as well as its general and specific treatments. DATA SOURCE A review was carried out in PubMed, Cochrane Library, and MRei consult databases, searching for articles related to the syndrome and published between 1995 and 2011. DATA SYNTHESIS Risk factors and the physiopathological mechanisms of the syndrome are discussed. The clinical presentation depends on the different factors involved. These are related to the etiology and physiopathology of the different forms of the disease. In addition to the measures used to allow for the decrease in pulmonary vascular resistance after birth, in some instances pulmonary vasodilators will be required. Although inhaled nitric oxide has proved effective, other vasodilators have been recently used, but clinical evidence is still lacking to demonstrate their benefits in the treatment of PPHN. CONCLUSIONS Despite recent technological advances and new physiopathological knowledge of this disease, mortality associated with PPHN remains at 10%. More clinical research and evidence-based experimental results are needed to prevent, treat, and reduce the morbidity/mortality associated with this neonatal syndrome.
Collapse
|
66
|
Delaney C, Gien J, Roe G, Isenberg N, Kailey J, Abman SH. Serotonin contributes to high pulmonary vascular tone in a sheep model of persistent pulmonary hypertension of the newborn. Am J Physiol Lung Cell Mol Physiol 2013; 304:L894-901. [PMID: 23605003 DOI: 10.1152/ajplung.00043.2013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although past studies demonstrate that altered serotonin (5-HT) signaling is present in adults with idiopathic pulmonary arterial hypertension, whether serotonin contributes to the pathogenesis of persistent pulmonary hypertension of the newborn (PPHN) is unknown. We hypothesized that 5-HT contributes to increased pulmonary vascular resistance (PVR) in a sheep model of PPHN and that selective 5-HT reuptake inhibitor (SSRI) treatment increases PVR in this model. We studied the hemodynamic effects of 5-HT, ketanserin (5-HT2A receptor antagonist), and sertraline, an SSRI, on pulmonary hemodynamics of the late gestation fetal sheep with PPHN caused by prolonged constriction of the ductus arteriosis. Brief intrapulmonary infusions of 5-HT increased PVR from 1.0 ± 0.07 (baseline) to 1.4 ± 0.22 mmHg/ml per minute of treatment (P < 0.05). Ketanserin decreased PVR from 1.1 ± 0.15 (baseline) to 0.82 ± 0.09 mmHg/ml per minute of treatment (P < 0.05). Sertraline increased PVR from 1.1 ± 0.17 (baseline) to 1.4 ± 0.17 mmHg/ml per minute of treatment (P = 0.01). In addition, we studied 5-HT production and activity in vitro in experimental PPHN. Compared with controls, pulmonary artery endothelial cells from fetal sheep with PPHN exhibited increased expression of tryptophan hydroxylase 1 and 5-HT production by twofold and 56%, respectively. Compared with controls, 5-HT2A R expression was increased in lung homogenates and pulmonary artery smooth muscle cell lysates by 35% and 32%, respectively. We concluded that increased 5-HT contributes to high PVR in experimental PPHN through activation of the 5-HT2A receptor and that SSRI infusion further increases PVR in this model.
Collapse
Affiliation(s)
- Cassidy Delaney
- Pediatric Heart Lung Center, Section of Neonatology, University of Colorado Denver, Aurora, CO 80045, USA.
| | | | | | | | | | | |
Collapse
|
67
|
Gupta V, Gupta N, Shaik IH, Mehvar R, Nozik-Grayck E, McMurtry IF, Oka M, Komatsu M, Ahsan F. Inhaled PLGA particles of prostaglandin E₁ ameliorate symptoms and progression of pulmonary hypertension at a reduced dosing frequency. Mol Pharm 2013; 10:1655-67. [PMID: 23485062 DOI: 10.1021/mp300426u] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
This study sought to investigate the efficacy of a noninvasive and long acting polymeric particle based formulation of prostaglandin E1 (PGE1), a potent pulmonary vasodilator, in alleviating the signs of pulmonary hypertension (PH) and reversing the biochemical changes that occur in the diseased lungs. PH rats, developed by a single subcutaneous injection of monocrotaline (MCT), were treated with two types of polymeric particles of PGE1, porous and nonporous, and intratracheal or intravenous plain PGE1. For chronic studies, rats received either intratracheal porous poly(lactic-co-glycolic acid) (PLGA) particles, once- or thrice-a-day, or plain PGE1 thrice-a-day for 10 days administered intratracheally or intravenously. The influence of formulations on disease progression was studied by measuring the mean pulmonary arterial pressure (MPAP), evaluating right ventricular hypertrophy and assessing various molecular and cellular makers including the degree of muscularization, platelet aggregation, matrix metalloproteinase-2 (MMP-2), and proliferating cell nuclear antigen (PCNA). Both plain PGE1 and large porous particles of PGE1 reduced MPAP and right ventricular hypertrophy (RVH) in rats that received the treatments for 10 days. Polymeric porous particles of PGE1 produced the same effects at a reduced dosing frequency compared to plain PGE1 and caused minimal off-target effects on systemic hemodynamics. Microscopic and immunohistochemical studies revealed that porous particles of PGE1 also reduced the degree of muscularization, von Willebrand factor (vWF), and PCNA expression in the lungs of PH rats. Overall, our study suggests that PGE1 loaded inhalable particulate formulations improve PH symptoms and arrest the progression of disease at a reduced dosing frequency compared to plain PGE1.
Collapse
Affiliation(s)
- Vivek Gupta
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S Coulter, Amarillo, Texas 79106, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Gien J, Tseng N, Seedorf G, Roe G, Abman SH. Endothelin-1 impairs angiogenesis in vitro through Rho-kinase activation after chronic intrauterine pulmonary hypertension in fetal sheep. Pediatr Res 2013; 73. [PMID: 23202724 PMCID: PMC3767851 DOI: 10.1038/pr.2012.177] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Endothelin-1 (ET-1) and Rho-kinase (ROCK) increase vascular tone in experimental persistent pulmonary hypertension of the newborn (PPHN). Whether ET-1 activates ROCK to decrease angiogenesis in the developing lung remains unknown. METHODS Proximal pulmonary artery endothelial cells (PAECs) were harvested from fetal sheep after partial ligation of the ductus arteriosus in utero (PPHN) and controls. Growth and tube formation were assessed after ET-1 treatment. The effect of ET-1 antagonism on tube formation was studied using ET-1 small interfering RNA (siRNA), ET-1 monoclonal antibodies (ET-1mAbs), BQ-123 (an ET(A) blocker), and bosentan (an ET(A)/ET(B) blocker). ET-1 gene and protein and ET(A)/ET(B) receptor protein expression were measured in normal and PPHN PAECs. ET-1-ROCK interactions were assessed by measuring ROCK activity after ET-1, ET-1 siRNA, and bosentan treatments, and tube formation with ET-1 and Y-27632 (ROCK inhibitor). RESULTS ET-1 did not affect growth but decreased tube formation in normal and PPHN PAECs. ET-1 protein and gene expression were increased and ET(B) receptor protein decreased in PPHN PAECs. ET-1 siRNA, ET-1mAbs, and bosentan, but not BQ-123, increased tube formation. ROCK activity was increased in PPHN PAECs and decreased with ET-1 siRNA and bosentan treatments. Y-27632 prevented the decrease in tube formation with ET-1. CONCLUSION ET-1 activation of ROCK impairs angiogenesis of fetal PAECs. Disruption of ET-1-ROCK interactions may increase vascular growth in PPHN.
Collapse
Affiliation(s)
- Jason Gien
- Department of Neonatology, University of Colorado School of Medicine, Aurora, Colorado, USA.
| | - Nancy Tseng
- Department of Neonatology University of Colorado School of Medicine, Aurora, CO, USA 80045
| | - Gregory Seedorf
- Department of Pulmonary Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA 80045
| | - Gates Roe
- Department of Neonatology University of Colorado School of Medicine, Aurora, CO, USA 80045
| | - Steven H. Abman
- Department of Pulmonary Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA 80045
| |
Collapse
|
69
|
Goncharova EA. mTOR and vascular remodeling in lung diseases: current challenges and therapeutic prospects. FASEB J 2013; 27:1796-807. [PMID: 23355268 DOI: 10.1096/fj.12-222224] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Mammalian target of rapamycin (mTOR) is a major regulator of cellular metabolism, proliferation, and survival that is implicated in various proliferative and metabolic diseases, including obesity, type 2 diabetes, hamartoma syndromes, and cancer. Emerging evidence suggests a potential critical role of mTOR signaling in pulmonary vascular remodeling. Remodeling of small pulmonary arteries due to increased proliferation, resistance to apoptosis, and altered metabolism of cells forming the pulmonary vascular wall is a key currently irreversible pathological feature of pulmonary hypertension, a progressive pulmonary vascular disorder with high morbidity and mortality. In addition to rare familial and idiopathic forms, pulmonary hypertension is also a life-threatening complication of several lung diseases associated with hypoxia. This review aims to summarize our current knowledge and recent advances in understanding the role of the mTOR pathway in pulmonary vascular remodeling, with a specific focus on the hypoxia component, a confirmed shared trigger of pulmonary hypertension in lung diseases. We also discuss the emerging role of mTOR as a promising therapeutic target and mTOR inhibitors as potential pharmacological approaches to treat pulmonary vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Elena A Goncharova
- University of Pennsylvania Perelman School of Medicine, Translational Research Laboratories, Rm. 1214, 125 South 31st St., Philadelphia, PA 19104, USA.
| |
Collapse
|
70
|
Vascular remodeling in pulmonary hypertension. J Mol Med (Berl) 2013; 91:297-309. [PMID: 23334338 DOI: 10.1007/s00109-013-0998-0] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 01/04/2013] [Accepted: 01/08/2013] [Indexed: 02/07/2023]
Abstract
Pulmonary hypertension is a complex, progressive condition arising from a variety of genetic and pathogenic causes. Patients present with a spectrum of histologic and pathophysiological features, likely reflecting the diversity in underlying pathogenesis. It is widely recognized that structural alterations in the vascular wall contribute to all forms of pulmonary hypertension. Features characteristic of the remodeled vasculature in patients with pulmonary hypertension include increased stiffening of the elastic proximal pulmonary arteries, thickening of the intimal and/or medial layer of muscular arteries, development of vaso-occlusive lesions, and the appearance of cells expressing smooth muscle-specific markers in normally non-muscular small diameter vessels, resulting from proliferation and migration of pulmonary arterial smooth muscle cells and cellular transdifferentiation. The development of several animal models of pulmonary hypertension has provided the means to explore the mechanistic underpinnings of pulmonary vascular remodeling, although none of the experimental models currently used entirely replicates the pulmonary arterial hypertension observed in patients. Herein, we provide an overview of the histological abnormalities observed in humans with pulmonary hypertension and in preclinical models and discuss insights gained regarding several key signaling pathways contributing to the remodeling process. In particular, we will focus on the roles of ion homeostasis, endothelin-1, serotonin, bone morphogenetic proteins, Rho kinase, and hypoxia-inducible factor 1 in pulmonary arterial smooth muscle and endothelial cells, highlighting areas of cross-talk between these pathways and potentials for therapeutic targeting.
Collapse
|
71
|
Schiavon M, Fadini GP, Lunardi F, Agostini C, Boscaro E, Calabrese F, Marulli G, Rea F. Increased tissue endothelial progenitor cells in end-stage lung diseases with pulmonary hypertension. J Heart Lung Transplant 2013; 31:1025-30. [PMID: 22884387 DOI: 10.1016/j.healun.2012.06.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 05/23/2012] [Accepted: 06/06/2012] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Diffuse lung diseases promote the development of vascular changes and pulmonary hypertension (PH). Endothelial progenitor cells (EPCs) seem to be involved in pulmonary vascular remodeling. We evaluated circulating and intra-pulmonary EPCs in end-stage lung diseases in relation to pulmonary arterial pressure (PAP). METHODS The study included 19 patients affected by different end-stage lung diseases, with or without PH. Six lung donors were considered as control group. EPCs were measured in blood samples taken at the time of transplant from pulmonary arteries and veins (by flow cytometry) as well as in lung specimen sections (by confocal microscopy) and expressed as percentage of total number of cells. RESULTS The amount of EPC in lung specimens was significantly different according to type of disease (p = 0.001). Specifically, a higher number of EPCs was detected in idiopathic pulmonary hypertension and idiopathic pulmonary fibrosis with high (> 25 mm Hg) mean PAP (p = 0.03 for both) compared with chronic obstructive pulmonary disease and control group. There was a direct correlation between intrapulmonary EPCs and PAP. According to receiver operating characteristic curve analysis, the presence of > 3% EPCs had a 91% sensitivity and 93% specificity in identifying high mean PAP. There were no differences in circulating arterial or venous EPCs among groups. CONCLUSIONS Intra-pulmonary EPCs are increased in lung diseases with high PAP, suggesting that EPCs may contribute to vascular remodeling in end-stage pulmonary disease.
Collapse
Affiliation(s)
- Marco Schiavon
- Department of Cardio-Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Blood AB, Terry MH, Merritt TA, Papamatheakis DG, Blood Q, Ross JM, Power GG, Longo LD, Wilson SM. Effect of chronic perinatal hypoxia on the role of rho-kinase in pulmonary artery contraction in newborn lambs. Am J Physiol Regul Integr Comp Physiol 2012; 304:R136-46. [PMID: 23152110 DOI: 10.1152/ajpregu.00126.2012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Exposure to chronic hypoxia during gestation predisposes infants to neonatal pulmonary hypertension, but the underlying mechanisms remain unclear. Here, we test the hypothesis that moderate continuous hypoxia during gestation causes changes in the rho-kinase pathway that persist in the newborn period, altering vessel tone and responsiveness. Lambs kept at 3,801 m above sea level during gestation and the first 2 wk of life were compared with those with gestation at low altitude. In vitro studies of isolated pulmonary arterial rings found a more forceful contraction in response to KCl and 5-HT in high-altitude compared with low-altitude lambs. There was no difference between the effects of blockers of various pathways of extracellular Ca(2+) entry in low- and high-altitude arteries. In contrast, inhibition of rho-kinase resulted in significantly greater attenuation of 5-HT constriction in high-altitude compared with low-altitude arteries. High-altitude lambs had higher baseline pulmonary artery pressures and greater elevations in pulmonary artery pressure during 15 min of acute hypoxia compared with low-altitude lambs. Despite evidence for an increased role for rho-kinase in high-altitude arteries, in vivo studies found no significant difference between the effects of rho-kinase inhibition on hypoxic pulmonary vasoconstriction in intact high-altitude and low-altitude lambs. We conclude that chronic hypoxia in utero results in increased vasopressor response to both acute hypoxia and serotonin, but that rho-kinase is involved only in the increased response to serotonin.
Collapse
Affiliation(s)
- Arlin B Blood
- Department of Pediatrics, Division of Neonatology, Loma Linda University School of Medicine, Loma Linda, CA 92373, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Jin Y, Chen B, Calvert TJ, Chicoine LG, Liu Y, Nelin LD. Chronic hypoxia decreases arterial and venous compliance in isolated perfused rat lungs: an effect that is reversed by exogenous L-arginine. Am J Physiol Heart Circ Physiol 2012; 304:H195-205. [PMID: 23103497 DOI: 10.1152/ajpheart.00188.2012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chronic hypoxia (CH)-induced pulmonary hypertension is characterized by vasoconstriction and vascular remodeling, leading to right ventricular dysfunction. Given the role of arterial compliance (C(a)) in right ventricular work, a decrease in C(a) would add to right ventricular work. Nitric oxide (NO) is a potent vasodilator made by NO synthases from L-arginine (L-Arg). However, little is known of the effect of L-Arg on vascular compliance (C(v)) in the lung. We hypothesized that exposure to CH would decrease C(a) and that this effect would be reversed by exogenous L-Arg. Sprague-Dawley rats were exposed to either normoxia or CH for 14 days; the lungs were then isolated and perfused. Vascular occlusions were performed and modeled using a three-compliance, two-resistor model. Pressure-flow curves were generated, and a distensible vessel model was used to estimate distensibility and a vascular resistance parameter (R(0)). Hypoxia resulted in the expected increase in arterial resistance (R(a)) as well as a decrease in both C(a) and C(v). L-Arg had little effect on R(a), C(a), or C(v) in isolated lungs from normoxic animals. L-Arg decreased R(a) in lungs from CH rats and redistributed compliance to approximately that found in normoxic lungs. CH increased R(0), and L-Arg reversed this increase in R(0). L-Arg increased exhaled NO, and inhibition of L-Arg uptake attenuated the L-Arg-induced increase in exhaled NO. These data demonstrate that the CH-induced decrease in C(a) was reversed by L-Arg, suggesting that L-Arg may improve CH-induced right ventricular dysfunction.
Collapse
Affiliation(s)
- Yi Jin
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | | | | | | | | | | |
Collapse
|
74
|
Fröhlich S, Boylan J, McLoughlin P. Hypoxia-induced inflammation in the lung: a potential therapeutic target in acute lung injury? Am J Respir Cell Mol Biol 2012; 48:271-9. [PMID: 23087053 DOI: 10.1165/rcmb.2012-0137tr] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Acute lung injury (ALI) is a severe form of hypoxic lung disease responsible for a large number of deaths worldwide. Despite recent advances in supportive care, no reduction in mortality has been evident since the introduction of a standard consensus definition almost two decades ago. New strategies are urgently required to help design effective therapies for this condition. A key pathological feature of ALI involves regional alveolar hypoxia. Because alveolar hypoxia in isolation, such as that encountered at high altitude, causes an inflammatory pulmonary phenotype in the absence of any other pathogenic stimuli, these regions may not be passive bystanders but may actually contribute to the pathogenesis and progression of lung injury. Unique transcriptional responses to hypoxia in the lung apparently allow it to express an inflammatory phenotype at levels of hypoxia that would not produce such a response in other organs. We will review recent advances in our understanding of these unique transcriptional responses to moderate levels of alveolar hypoxia, which may provide new insights into the pathogenesis of ALI.
Collapse
Affiliation(s)
- Stephen Fröhlich
- Department of Anaesthesia and Intensive Care, St. Vincent's University Hospital, Dublin 4, Ireland.
| | | | | |
Collapse
|
75
|
Singh GK, Levy PT, Holland MR, Hamvas A. Novel methods for assessment of right heart structure and function in pulmonary hypertension. Clin Perinatol 2012; 39:685-701. [PMID: 22954276 DOI: 10.1016/j.clp.2012.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Long-term increases in pulmonary vascular resistance and pulmonary arterial pressure resulting from structural alterations and abnormal vasoreactivity of the pulmonary vasculature may lead to right ventricular (RV) remodeling. Conventional methods of assessment of RV structure and function do not provide sensitive markers of RV remodeling for prognostic information. Advances in cardiac imaging have provided the capability to obtain quantitative information on the RV structure and function. This article reviews the clinical conditions that result in PH and discusses the novel and emerging methods for the assessment of right heart structure and function in PH in infants and children.
Collapse
Affiliation(s)
- Gautam K Singh
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA.
| | | | | | | |
Collapse
|
76
|
Li CB, Li XX, Chen YG, Gao HQ, Bao MC, Zhang J, Bu PL, Zhang Y, Ji XP. Simvastatin exerts cardioprotective effects and inhibits the activity of Rho-associated protein kinase in rats with metabolic syndrome. Clin Exp Pharmacol Physiol 2012; 39:759-64. [PMID: 22670687 DOI: 10.1111/j.1440-1681.2012.05730.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
| | - Xiao-Xing Li
- Department of Geriatrics; Shandong University Qilu Hospital; Jinan; Shandong; China
| | | | - Hai-Qing Gao
- Department of Geriatrics; Shandong University Qilu Hospital; Jinan; Shandong; China
| | - Mei-Cheng Bao
- Department of Internal Medicine; Jinan Second People's Hospital; Jinan; Shandong; China
| | - Juan Zhang
- The Key Laboratory of Cardiovascular Remodelling and Function Research; Chinese Ministry of Education and Chinese Ministry of Public Health; Jinan; Shandong; China
| | - Pei-Li Bu
- The Key Laboratory of Cardiovascular Remodelling and Function Research; Chinese Ministry of Education and Chinese Ministry of Public Health; Jinan; Shandong; China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodelling and Function Research; Chinese Ministry of Education and Chinese Ministry of Public Health; Jinan; Shandong; China
| | - Xiao-Ping Ji
- The Key Laboratory of Cardiovascular Remodelling and Function Research; Chinese Ministry of Education and Chinese Ministry of Public Health; Jinan; Shandong; China
| |
Collapse
|
77
|
Buravkova LB, Andreeva ER, Grigoriev AI. The impact of oxygen in physiological regulation of human multipotent mesenchymal cell functions. ACTA ACUST UNITED AC 2012. [DOI: 10.1134/s0362119712040032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
78
|
Regulation of S100A4 expression via the JAK2–STAT3 pathway in rhomboid-phenotype pulmonary arterial smooth muscle cells exposure to hypoxia. Int J Biochem Cell Biol 2012; 44:1337-45. [DOI: 10.1016/j.biocel.2012.04.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 04/02/2012] [Accepted: 04/22/2012] [Indexed: 01/27/2023]
|
79
|
Firth AL, Choi IW, Park WS. Animal models of pulmonary hypertension: Rho kinase inhibition. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2012; 109:67-75. [PMID: 22713173 DOI: 10.1016/j.pbiomolbio.2012.05.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 05/17/2012] [Accepted: 05/21/2012] [Indexed: 10/28/2022]
Abstract
Pulmonary Hypertension is a terminology encompassing a range of etiologically different pulmonary vascular diseases. The most common is that termed pulmonary arterial hypertension or PAH; a rare but often fatal disease characterized by a mean pulmonary arterial pressure of >25 mmHg. PAH is associated with a complex etiology highlighted by core characteristics of increased pulmonary vascular resistance and elevation of mean pulmonary artery pressure. When sustained, pulmonary vascular remodeling occurs and eventually patients pass away due to right heart failure. Hypoxic pulmonary vasoconstriction is an early event occurring in pulmonary hypertension due to chronic exposure to hypoxia. While the underlying mechanisms of hypoxic pulmonary vasoconstriction may be controversial, a role for RhoA/Rho kinase mediated regulation of intracellular Ca(2+) has been recently identified. Further study suggests that RhoA may have an integral role in other pathophysiological processes such as cell proliferation and migration occurring in all forms of PH. Indeed Rho proteins are known to play essential roles in actin cytoskeleton organization in all eukaryotic cells and thus Rho and Rho-GTPases are implicated in fundamental cellular processes such as cellular proliferation, migration, adhesion, apoptosis and gene expression. This review focuses on providing an overview of the role of RhoA/Rho kinase in currently available animal models of pulmonary hypertension.
Collapse
Affiliation(s)
- Amy L Firth
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | | |
Collapse
|
80
|
Zhu D, Ran Y. Role of 15-lipoxygenase/15-hydroxyeicosatetraenoic acid in hypoxia-induced pulmonary hypertension. J Physiol Sci 2012; 62:163-72. [PMID: 22331435 PMCID: PMC10717549 DOI: 10.1007/s12576-012-0196-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 01/25/2012] [Indexed: 12/01/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease with a complex aetiology characterized by elevated pulmonary artery resistance, which leads to right heart ventricular afterload and ultimately progressing to right ventricular failure and often death. In addition to other factors, metabolites of arachidonic acid cascade play an important role in the pulmonary vasculature, and disruption of signaling pathways of arachidonic acid plays a central role in the pathogenesis of PAH. 15-Lipoxygenase (15-LO) is upregulated in pulmonary artery endothelial cells and smooth muscle cells of PAH patients, and its metabolite 15-hydroxyeicosatetraenoic acid (15-HETE) in particular seems to play a central role in the contractile machinery, and in the initiation and propagation of cell proliferation via its effects on signal pathways, mitogens, and cell cycle components. Here, we focus on our important research into the role played by 15-LO/15-HETE, which promotes a proliferative, antiapoptotic, and vasoconstrictive physiological milieu leading to hypoxic pulmonary hypertension.
Collapse
Affiliation(s)
- Daling Zhu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, Heilongjiang, People's Republic of China.
| | | |
Collapse
|
81
|
Cahill E, Rowan SC, Sands M, Banahan M, Ryan D, Howell K, McLoughlin P. The pathophysiological basis of chronic hypoxic pulmonary hypertension in the mouse: vasoconstrictor and structural mechanisms contribute equally. Exp Physiol 2012; 97:796-806. [DOI: 10.1113/expphysiol.2012.065474] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
82
|
Acetylsalicylic acid regulates overexpressed small GTPase RhoA in vascular smooth muscle cells through prevention of new synthesis and enhancement of protein degradation. Biosci Rep 2012; 32:153-60. [PMID: 21756248 DOI: 10.1042/bsr20110050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
RhoA has been shown to play a major role in vascular processes and acetylsalicylic acid (aspirin) is known to exert a cytoprotective effect via multiple mechanisms. In the present study, we aimed at investigating the effect of aspirin on RhoA expression under a stress state in rat VSMCs (vascular smooth muscle cells) and the underlying mechanisms. The expression of iNOS (inducible nitric oxide synthase) and iNOS activity as well as NO concentration was significantly promoted by LPS (lipopolysaccharide) accompanying the elevation of RhoA expression, which was blocked by the addition of the iNOS inhibitor L-NIL [L-N6-(1-iminoethyl)lysine dihydrochloride]. Aspirin (30 μM) significantly attenuated the elevation of RhoA, while indomethacin and salicylate had no similar effect. The sGC (soluble guanylate cyclase) inhibitor ODQ (1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one) showed the same effect as aspirin in down-regulating RhoA but was reversed by the addition of the cGMP analogue 8-Br-PET-cGMP (β-phenyl-1,N2-ethano-8-bromoguanosine 3',5'-cyclic monophosphorothioate). 8-Br-PET-cGMP solely enhanced the RhoA expression that was abrogated by preincubation with aspirin. Degradation analysis indicated that aspirin enhanced the protein degradation rate of RhoA and GDP-bound RhoA seemed to be more susceptible to aspirin-enhanced degradation compared with the GTP-bound form. Our results indicate that aspirin attenuates the LPS-induced overexpression of RhoA both by inhibiting new synthesis and accelerating protein degradation, which may help elucidate the multiple beneficial effects of aspirin.
Collapse
|
83
|
Cahill E, Costello CM, Rowan SC, Harkin S, Howell K, Leonard MO, Southwood M, Cummins EP, Fitzpatrick SF, Taylor CT, Morrell NW, Martin F, McLoughlin P. Gremlin plays a key role in the pathogenesis of pulmonary hypertension. Circulation 2012; 125:920-30. [PMID: 22247494 DOI: 10.1161/circulationaha.111.038125] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pulmonary hypertension occurs in chronic hypoxic lung diseases, significantly worsening morbidity and mortality. The important role of altered bone morphogenetic protein (BMP) signaling in pulmonary hypertension was first suspected after the identification of heterozygous BMP receptor mutations as the underlying defect in the rare heritable form of pulmonary arterial hypertension. Subsequently, it was demonstrated that BMP signaling was also reduced in common forms of pulmonary hypertension, including hypoxic pulmonary hypertension; however, the mechanism of this reduction has not previously been elucidated. METHODS AND RESULTS Expression of 2 BMP antagonists, gremlin 1 and gremlin 2, was higher in the lung than in other organs, and gremlin 1 was further increased in the walls of small intrapulmonary vessels of mice during the development of hypoxic pulmonary hypertension. Hypoxia stimulated gremlin secretion from human pulmonary microvascular endothelial cells in vitro, which inhibited endothelial BMP signaling and BMP-stimulated endothelial repair. Haplodeficiency of gremlin 1 augmented BMP signaling in the hypoxic mouse lung and reduced pulmonary vascular resistance by attenuating vascular remodeling. Furthermore, gremlin was increased in the walls of small intrapulmonary vessels in idiopathic pulmonary arterial hypertension and the rare heritable form of pulmonary arterial hypertension in a distribution suggesting endothelial localization. CONCLUSIONS These findings demonstrate a central role for increased gremlin in hypoxia-induced pulmonary vascular remodeling and the increased pulmonary vascular resistance in hypoxic pulmonary hypertension. High levels of basal gremlin expression in the lung may account for the unique vulnerability of the pulmonary circulation to heterozygous mutations of BMP type 2 receptor in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Edwina Cahill
- University College Dublin, School of Medicine and Medical Sciences, Belfield, Dublin 4, Ireland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Abstract
It has been known for more than 60 years, and suspected for over 100, that alveolar hypoxia causes pulmonary vasoconstriction by means of mechanisms local to the lung. For the last 20 years, it has been clear that the essential sensor, transduction, and effector mechanisms responsible for hypoxic pulmonary vasoconstriction (HPV) reside in the pulmonary arterial smooth muscle cell. The main focus of this review is the cellular and molecular work performed to clarify these intrinsic mechanisms and to determine how they are facilitated and inhibited by the extrinsic influences of other cells. Because the interaction of intrinsic and extrinsic mechanisms is likely to shape expression of HPV in vivo, we relate results obtained in cells to HPV in more intact preparations, such as intact and isolated lungs and isolated pulmonary vessels. Finally, we evaluate evidence regarding the contribution of HPV to the physiological and pathophysiological processes involved in the transition from fetal to neonatal life, pulmonary gas exchange, high-altitude pulmonary edema, and pulmonary hypertension. Although understanding of HPV has advanced significantly, major areas of ignorance and uncertainty await resolution.
Collapse
Affiliation(s)
- J T Sylvester
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, The Johns Hopkins University School ofMedicine, Baltimore, Maryland, USA.
| | | | | | | |
Collapse
|
85
|
Lammers S, Scott D, Hunter K, Tan W, Shandas R, Stenmark KR. Mechanics and Function of the Pulmonary Vasculature: Implications for Pulmonary Vascular Disease and Right Ventricular Function. Compr Physiol 2012; 2:295-319. [PMID: 23487595 DOI: 10.1002/cphy.c100070] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The relationship between cardiac function and the afterload against which the heart muscle must work to circulate blood throughout the pulmonary circulation is defined by a complex interaction between many coupled system parameters. These parameters range broadly and incorporate system effects originating primarily from three distinct locations: input power from the heart, hydraulic impedance from the large conduit pulmonary arteries, and hydraulic resistance from the more distal microcirculation. These organ systems are not independent, but rather, form a coupled system in which a change to any individual parameter affects all other system parameters. The result is a highly nonlinear system which requires not only detailed study of each specific component and the effect of disease on their specific function, but also requires study of the interconnected relationship between the microcirculation, the conduit arteries, and the heart in response to age and disease. Here, we investigate systems-level changes associated with pulmonary hypertensive disease progression in an effort to better understand this coupled relationship.
Collapse
Affiliation(s)
- Steven Lammers
- Department of Cardiovascular Pulmonary Research, University of Colorado Denver, Aurora, Colorado ; Department of Bioengineering, University of Colorado Denver, Aurora, Colorado
| | | | | | | | | | | |
Collapse
|
86
|
Fang X, Ueno M, Yamashita T, Ikuno Y. RhoA Activation and Effect of Rho-kinase Inhibitor in the Development of Retinal Neovascularization in a Mouse Model of Oxygen-induced Retinopathy. Curr Eye Res 2011; 36:1028-36. [DOI: 10.3109/02713683.2011.593110] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
87
|
Ma C, Li Y, Ma J, Liu Y, Li Q, Niu S, Shen Z, Zhang L, Pan Z, Zhu D. Key Role of 15-Lipoxygenase/15-Hydroxyeicosatetraenoic Acid in Pulmonary Vascular Remodeling and Vascular Angiogenesis Associated With Hypoxic Pulmonary Hypertension. Hypertension 2011; 58:679-88. [DOI: 10.1161/hypertensionaha.111.171561] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have found that 15-hydroxyeicosatetraenoic acid (15-HETE) induced by hypoxia was an important mediator in the regulation of hypoxic pulmonary hypertension, including the pulmonary vasoconstriction and remodeling. However, the underlying mechanisms of the remodeling induced by 15-HETE are poorly understood. In this study, we performed immunohistochemistry, pulmonary artery endothelial cells migration and tube formation, pulmonary artery smooth muscle cells bromodeoxyuridine incorporation, and cell cycle analysis to determine the role of 15-HETE in hypoxia-induced pulmonary vascular remodeling. We found that hypoxia induced pulmonary vascular medial hypertrophy and intimal endothelial cells migration and angiogenesis, which were mediated by 15-HETE. Moreover, 15-HETE regulated the cell cycle progression and made more smooth muscle cells from the G
0
/G
1
phase to the G
2
/M+S phase and enhanced the microtubule formation in cell nucleus. In addition, we found that the Rho-kinase pathway was involved in 15-HETE–induced endothelial cells tube formation and migration and smooth muscle cell proliferation. Together, these results show that 15-HETE mediates hypoxia-induced pulmonary vascular remodeling and stimulates angiogenesis via the Rho-kinase pathway.
Collapse
Affiliation(s)
- Cui Ma
- From the Department of Biopharmaceutical Sciences (C.M., Y. Li, J.M., Y. Liu, Q.L., S.N., L.Z., Z.P., D.Z.), College of Pharmacy, Harbin Medical University, Nangang District, Harbin, Heilongjiang, People's Republic of China; Bio-pharmaceutical Key Laboratory of Heilongjiang Province (D.Z.), Harbin, People's Republic of China; Department of Pharmacology (Z.S.), Harbin Medical University-Daqing, Daqing, Heilongjiang Province, People's Republic of China
| | - Yaqian Li
- From the Department of Biopharmaceutical Sciences (C.M., Y. Li, J.M., Y. Liu, Q.L., S.N., L.Z., Z.P., D.Z.), College of Pharmacy, Harbin Medical University, Nangang District, Harbin, Heilongjiang, People's Republic of China; Bio-pharmaceutical Key Laboratory of Heilongjiang Province (D.Z.), Harbin, People's Republic of China; Department of Pharmacology (Z.S.), Harbin Medical University-Daqing, Daqing, Heilongjiang Province, People's Republic of China
| | - Jun Ma
- From the Department of Biopharmaceutical Sciences (C.M., Y. Li, J.M., Y. Liu, Q.L., S.N., L.Z., Z.P., D.Z.), College of Pharmacy, Harbin Medical University, Nangang District, Harbin, Heilongjiang, People's Republic of China; Bio-pharmaceutical Key Laboratory of Heilongjiang Province (D.Z.), Harbin, People's Republic of China; Department of Pharmacology (Z.S.), Harbin Medical University-Daqing, Daqing, Heilongjiang Province, People's Republic of China
| | - Yun Liu
- From the Department of Biopharmaceutical Sciences (C.M., Y. Li, J.M., Y. Liu, Q.L., S.N., L.Z., Z.P., D.Z.), College of Pharmacy, Harbin Medical University, Nangang District, Harbin, Heilongjiang, People's Republic of China; Bio-pharmaceutical Key Laboratory of Heilongjiang Province (D.Z.), Harbin, People's Republic of China; Department of Pharmacology (Z.S.), Harbin Medical University-Daqing, Daqing, Heilongjiang Province, People's Republic of China
| | - Qian Li
- From the Department of Biopharmaceutical Sciences (C.M., Y. Li, J.M., Y. Liu, Q.L., S.N., L.Z., Z.P., D.Z.), College of Pharmacy, Harbin Medical University, Nangang District, Harbin, Heilongjiang, People's Republic of China; Bio-pharmaceutical Key Laboratory of Heilongjiang Province (D.Z.), Harbin, People's Republic of China; Department of Pharmacology (Z.S.), Harbin Medical University-Daqing, Daqing, Heilongjiang Province, People's Republic of China
| | - Shengpan Niu
- From the Department of Biopharmaceutical Sciences (C.M., Y. Li, J.M., Y. Liu, Q.L., S.N., L.Z., Z.P., D.Z.), College of Pharmacy, Harbin Medical University, Nangang District, Harbin, Heilongjiang, People's Republic of China; Bio-pharmaceutical Key Laboratory of Heilongjiang Province (D.Z.), Harbin, People's Republic of China; Department of Pharmacology (Z.S.), Harbin Medical University-Daqing, Daqing, Heilongjiang Province, People's Republic of China
| | - Zhiying Shen
- From the Department of Biopharmaceutical Sciences (C.M., Y. Li, J.M., Y. Liu, Q.L., S.N., L.Z., Z.P., D.Z.), College of Pharmacy, Harbin Medical University, Nangang District, Harbin, Heilongjiang, People's Republic of China; Bio-pharmaceutical Key Laboratory of Heilongjiang Province (D.Z.), Harbin, People's Republic of China; Department of Pharmacology (Z.S.), Harbin Medical University-Daqing, Daqing, Heilongjiang Province, People's Republic of China
| | - Lei Zhang
- From the Department of Biopharmaceutical Sciences (C.M., Y. Li, J.M., Y. Liu, Q.L., S.N., L.Z., Z.P., D.Z.), College of Pharmacy, Harbin Medical University, Nangang District, Harbin, Heilongjiang, People's Republic of China; Bio-pharmaceutical Key Laboratory of Heilongjiang Province (D.Z.), Harbin, People's Republic of China; Department of Pharmacology (Z.S.), Harbin Medical University-Daqing, Daqing, Heilongjiang Province, People's Republic of China
| | - Zhenwei Pan
- From the Department of Biopharmaceutical Sciences (C.M., Y. Li, J.M., Y. Liu, Q.L., S.N., L.Z., Z.P., D.Z.), College of Pharmacy, Harbin Medical University, Nangang District, Harbin, Heilongjiang, People's Republic of China; Bio-pharmaceutical Key Laboratory of Heilongjiang Province (D.Z.), Harbin, People's Republic of China; Department of Pharmacology (Z.S.), Harbin Medical University-Daqing, Daqing, Heilongjiang Province, People's Republic of China
| | - Daling Zhu
- From the Department of Biopharmaceutical Sciences (C.M., Y. Li, J.M., Y. Liu, Q.L., S.N., L.Z., Z.P., D.Z.), College of Pharmacy, Harbin Medical University, Nangang District, Harbin, Heilongjiang, People's Republic of China; Bio-pharmaceutical Key Laboratory of Heilongjiang Province (D.Z.), Harbin, People's Republic of China; Department of Pharmacology (Z.S.), Harbin Medical University-Daqing, Daqing, Heilongjiang Province, People's Republic of China
| |
Collapse
|
88
|
Delaney C, Gien J, Grover TR, Roe G, Abman SH. Pulmonary vascular effects of serotonin and selective serotonin reuptake inhibitors in the late-gestation ovine fetus. Am J Physiol Lung Cell Mol Physiol 2011; 301:L937-44. [PMID: 21908589 DOI: 10.1152/ajplung.00198.2011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Maternal use of selective serotonin (5-HT) reuptake inhibitors (SSRIs) is associated with an increased risk for persistent pulmonary hypertension of the newborn (PPHN), but little is known about 5-HT signaling in the developing lung. We hypothesize that 5-HT plays a key role in maintaining high pulmonary vascular resistance (PVR) in the fetus and that fetal exposure to SSRIs increases 5-HT activity and causes pulmonary hypertension. We studied the hemodynamic effects of 5-HT, 5-HT receptor antagonists, and SSRIs in chronically prepared fetal sheep. Brief infusions of 5-HT (3-20 μg) increased PVR in a dose-related fashion. Ketanserin, a 5-HT 2A receptor antagonist, caused pulmonary vasodilation and inhibited 5-HT-induced pulmonary vasoconstriction. In contrast, intrapulmonary infusions of GR127945 and SB206553, 5-HT 1B and 5-HT 2B receptor antagonists, respectively, had no effect on basal PVR or 5-HT-induced vasoconstriction. Pretreatment with fasudil, a Rho kinase inhibitor, blunted the effects of 5-HT infusion. Brief infusions of the SSRIs, sertraline and fluoxetine, caused potent and sustained elevations of PVR, which was sustained for over 60 min after the infusion. SSRI-induced pulmonary vasoconstriction was reversed by infusion of ketanserin and did not affect the acute vasodilator effects of acetylcholine. We conclude that 5-HT causes pulmonary vasoconstriction, contributes to maintenance of high PVR in the normal fetus through stimulation of 5-HT 2A receptors and Rho kinase activation, and mediates the hypertensive effects of SSRIs. We speculate that prolonged exposure to SSRIs can induce PPHN through direct effects on the fetal pulmonary circulation.
Collapse
|
89
|
Klemm DJ, Majka SM, Crossno JT, Psilas JC, Reusch JE, Garat CV. Reduction of reactive oxygen species prevents hypoxia-induced CREB depletion in pulmonary artery smooth muscle cells. J Cardiovasc Pharmacol 2011; 58:181-91. [PMID: 21562428 PMCID: PMC3155008 DOI: 10.1097/fjc.0b013e31821f2773] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Hypoxia-induced pulmonary arterial hypertension (PAH) is a deadly disease characterized by progressive remodeling and persistent vasoconstriction of the pulmonary arterial system. Remodeling of the pulmonary artery (PA) involves smooth muscle cell (SMC) proliferation, hypertrophy, migration, and elevated extracellular matrix (ECM) production elicited by mitogens and oxidants produced in response to hypoxic insult. We previously reported that the transcription factor cAMP response element binding protein (CREB) is depleted in medial PA SMCs in remodeled, hypertensive vessels in rats or calves exposed to chronic hypoxia. In culture, CREB loss can be induced in PA SMCs by exogenous oxidants or platelet-derived growth factor. Forced depletion of CREB with small interfering RNA (siRNA) in PA SMCs is sufficient to induce their proliferation, hypertrophy, migration, dedifferentiation, and ECM production. This suggests that oxidant and/or mitogen-induced loss of CREB in medial SMCs is, in part, responsible for PA thickening. Here, we tested whether oxidant scavengers could prevent the loss of CREB in PA SMCs and inhibit SMC proliferation, migration, and ECM production using in vitro and in vivo models. Exposure of PA SMCs to hypoxia induced hydrogen peroxide (H2O2) production and loss of CREB. Treatment of SMCs with exogenous H2O2 or a second oxidant, Sin-1, elicited CREB depletion under normoxic conditions. Exogenous H2O2 also induced SMC proliferation, migration, and increased elastin levels as did forced depletion of CREB. In vivo, hypoxia-induced thickening of the PA wall was suppressed by the superoxide dismutase mimetic, Tempol, which also prevented the loss of CREB in medial SMCs. Tempol also reduced hypoxia-induced SMC proliferation and elastin deposition in the PA. The data indicate that CREB levels in the arterial wall are regulated in part by oxidants produced in response to hypoxia and that CREB plays a crucial role in regulating SMC phenotype and PA remodeling.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cell Culture Techniques
- Cell Hypoxia/drug effects
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors
- Cyclic AMP Response Element-Binding Protein/biosynthesis
- Free Radical Scavengers/pharmacology
- Hydrogen Peroxide/metabolism
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/metabolism
- Hypoxia/complications
- Hypoxia/metabolism
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Pulmonary Artery/cytology
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- RNA, Small Interfering/pharmacology
- Rats
- Rats, Inbred WKY
Collapse
Affiliation(s)
- Dwight J. Klemm
- Cardiovascular Pulmonary Research, University of Colorado Denver, Aurora, CO 80045
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Susan M. Majka
- Division of Cardiology, University of Colorado Denver, Aurora, CO 80045
| | - Joseph T. Crossno
- Cardiovascular Pulmonary Research, University of Colorado Denver, Aurora, CO 80045
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045
| | - John C. Psilas
- Cardiovascular Pulmonary Research, University of Colorado Denver, Aurora, CO 80045
| | - Jane E.B. Reusch
- Division of Endocrinology, University of Colorado Denver, Aurora, CO 80045
| | - Chrystelle V. Garat
- Cardiovascular Pulmonary Research, University of Colorado Denver, Aurora, CO 80045
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045
| |
Collapse
|
90
|
Nisbet RE, Sutliff RL, Hart CM. The role of peroxisome proliferator-activated receptors in pulmonary vascular disease. PPAR Res 2011; 2007:18797. [PMID: 17710111 PMCID: PMC1940049 DOI: 10.1155/2007/18797] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2007] [Accepted: 04/30/2007] [Indexed: 02/07/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors belonging to the nuclear hormone
receptor superfamily that regulate diverse physiological processes ranging from lipogenesis to inflammation. Recent evidence has
established potential roles of PPARs in both systemic and pulmonary vascular disease and function. Existing treatment strategies
for pulmonary hypertension, the most common manifestation of pulmonary vascular disease, are limited by an incomplete
understanding of the underlying disease pathogenesis and lack of efficacy indicating an urgent need for new approaches to treat
this disorder. Derangements in pulmonary endothelial-derived mediators and endothelial dysfunction have been shown to play a
pivotal role in pulmonary hypertension pathogenesis. Therefore, the following review will focus on selected mediators implicated
in pulmonary vascular dysfunction and evidence that PPARs, in particular PPARγ, participate in their regulation and may provide
a potential novel therapeutic target for the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
- Rachel E. Nisbet
- Department of Medicine, Emory University, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
- *Rachel E. Nisbet:
| | - Roy L. Sutliff
- Department of Medicine, Emory University, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
| | - C. Michael Hart
- Department of Medicine, Emory University, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
| |
Collapse
|
91
|
Hunter KS, Lammers SR, Shandas R. Pulmonary vascular stiffness: measurement, modeling, and implications in normal and hypertensive pulmonary circulations. Compr Physiol 2011; 1:1413-35. [PMID: 23733649 PMCID: PMC4113421 DOI: 10.1002/cphy.c100005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This article introduces the concept of pulmonary vascular stiffness, discusses its increasingly recognized importance as a diagnostic marker in the evaluation of pulmonary vascular disease, and describes methods to measure and model it clinically, experimentally, and computationally. It begins with a description of systems-level methods to evaluate pulmonary vascular compliance and recent clinical efforts in applying such techniques to better predict patient outcomes in pulmonary arterial hypertension. It then progresses from the systems-level to the local level, discusses proposed methods by which upstream pulmonary vessels increase in stiffness, introduces concepts around vascular mechanics, and concludes by describing recent work incorporating advanced numerical methods to more thoroughly evaluate changes in local mechanical properties of pulmonary arteries.
Collapse
Affiliation(s)
- Kendall S. Hunter
- Department of Bioengineering, University of Colorado at Denver Anschutz Medical Campus (UCD-AMC), Aurora, Colorado
- Division of Cardiology, Department of Pediatrics, The Children’s Hospital of Denver, UCD-AMC, Aurora, Colorado
| | - Steven R. Lammers
- Department of Bioengineering, University of Colorado at Denver Anschutz Medical Campus (UCD-AMC), Aurora, Colorado
- Cardiovascular Pulmonary (CVP) Research Laboratory, UCD-AMC, Aurora, Colorado
| | - Robin Shandas
- Department of Bioengineering, University of Colorado at Denver Anschutz Medical Campus (UCD-AMC), Aurora, Colorado
- Division of Cardiology, Department of Pediatrics, The Children’s Hospital of Denver, UCD-AMC, Aurora, Colorado
- Department of Surgery, UCD-AMC, Aurora, Colorado
| |
Collapse
|
92
|
Wagner EM, Jenkins J, Perino MG, Sukkar A, Mitzner W. Lung and vascular function during chronic severe pulmonary ischemia. J Appl Physiol (1985) 2011; 110:538-44. [PMID: 21148340 PMCID: PMC3043788 DOI: 10.1152/japplphysiol.01308.2010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 12/08/2010] [Indexed: 12/20/2022] Open
Abstract
Bronchial vascular angiogenesis takes place in a variety of lung inflammatory conditions such as asthma, cystic fibrosis, lung cancer, and chronic pulmonary thromboembolic disease. However, it is unclear whether neovascularization is predominantly appropriate and preserves lung tissue or whether it contributes further to lung pathology through edema formation and inflammation. In the present study we examined airway and lung parenchymal function 14 days after left pulmonary artery ligation. In rats as well as higher mammals, severe pulmonary ischemia results in bronchial vascular proliferation. Using labeled microspheres, we demonstrated an 18-fold increase in systemic blood flow to the ischemic left lung. Additionally, vascular remodeling extended to the tracheal venules, which showed an average 28% increase in venular diameter. Despite this increase in vascularity, airways resistance was not altered nor was methacholine responsiveness. Since these measurements include the entire lung, we suggest that the normal right lung, which represented 78% of the total lung, obscured the ability to detect a change. When functional indexes such as diffusing capacity, in situ lung volume, and vascular permeability of the left lung could be separated from right lung, significant changes were observed. Thus when comparing average left lung values of rats 14 days after left pulmonary artery ligation to left lungs of rats undergoing sham surgery, diffusing capacity of the left lung decreased by 72%, left lung volume decreased by 38%, and the vascular permeability to protein increased by 58%. No significant differences in inflammatory cell recruitment were observed, suggesting that acute ischemic inflammation had resolved. We conclude that despite the preservation of lung tissue, the proliferating bronchial neovasculature may contribute to a sustained decrement in pulmonary function.
Collapse
Affiliation(s)
- Elizabeth M Wagner
- Johns Hopkins Asthma and Allergy Center, Div. of Pulmonary and Critical Care Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA.
| | | | | | | | | |
Collapse
|
93
|
Wideman RF, Hamal KR. Idiopathic pulmonary arterial hypertension: an avian model for plexogenic arteriopathy and serotonergic vasoconstriction. J Pharmacol Toxicol Methods 2011; 63:283-95. [PMID: 21277983 DOI: 10.1016/j.vascn.2011.01.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 12/15/2010] [Accepted: 01/18/2011] [Indexed: 01/15/2023]
Abstract
Idiopathic pulmonary arterial hypertension (IPAH) is a disease of unknown cause that is characterized by elevated pulmonary arterial pressure and pulmonary vascular resistance attributable to vasoconstriction and vascular remodeling of small pulmonary arteries. Vascular remodeling includes hypertrophy and hyperplasia of smooth muscle (medial hypertrophy) accompanied in up to 80% of the cases by the formation of occlusive plexiform lesions (plexogenic arteriopathy). Patients tend to be unresponsive to vasodilator therapy and have a poor prognosis for survival when plexogenic arteriopathy progressively obstructs their pulmonary arteries. Research is needed to understand and treat plexogenic arteriopathy, but advances have been hindered by the absence of spontaneously developing lesions in existing laboratory animal models. Young domestic fowl bred for meat production (broiler chickens, broilers) spontaneously develop IPAH accompanied by semi-occlusive endothelial proliferation that progresses into fully developed plexiform lesions. Plexiform lesions develop in both female and male broilers, and lesion incidences (lung sections with lesions/lung sections examined) averaged approximately 40% in 8 to 52 week old birds. Plexiform lesions formed distal to branch points in muscular interparabronchial pulmonary arteries, and were associated with perivascular mononuclear cell infiltrates. Serotonin (5-hydroxytryptamine, 5-HT) is a potent vasoconstrictor and mitogen known to stimulate vascular endothelial and smooth muscle cell proliferation. Serotonin has been directly linked to the pathogenesis of IPAH in humans, including IPAH linked to serotonergic anorexigens that trigger the formation of plexiform lesions indistinguishable from those observed in primary IPAH triggered by other causes. Serotonin also plays a major role in the susceptibility of broilers to IPAH. This avian model of spontaneous IPAH constitutes a new animal model for biomedical research focused on the pathogenesis of IPAH and plexogenic arteriopathy.
Collapse
Affiliation(s)
- Robert F Wideman
- Center of Excellence for Poultry Science, Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA.
| | | |
Collapse
|
94
|
Sands M, Howell K, Costello CM, McLoughlin P. Placenta growth factor and vascular endothelial growth factor B expression in the hypoxic lung. Respir Res 2011; 12:17. [PMID: 21266048 PMCID: PMC3040134 DOI: 10.1186/1465-9921-12-17] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 01/25/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chronic alveolar hypoxia, due to residence at high altitude or chronic obstructive lung diseases, leads to pulmonary hypertension, which may be further complicated by right heart failure, increasing morbidity and mortality. In the non-diseased lung, angiogenesis occurs in chronic hypoxia and may act in a protective, adaptive manner. To date, little is known about the behaviour of individual vascular endothelial growth factor (VEGF) family ligands in hypoxia-induced pulmonary angiogenesis. The aim of this study was to examine the expression of placenta growth factor (PlGF) and VEGFB during the development of hypoxic pulmonary angiogenesis and their functional effects on the pulmonary endothelium. METHODS Male Sprague Dawley rats were exposed to conditions of normoxia (21% O2) or hypoxia (10% O2) for 1-21 days. Stereological analysis of vascular structure, real-time PCR analysis of vascular endothelial growth factor A (VEGFA), VEGFB, placenta growth factor (PlGF), VEGF receptor 1 (VEGFR1) and VEGFR2, immunohistochemistry and western blots were completed. The effects of VEGF ligands on human pulmonary microvascular endothelial cells were determined using a wound-healing assay. RESULTS Typical vascular remodelling and angiogenesis were observed in the hypoxic lung. PlGF and VEGFB mRNA expression were significantly increased in the hypoxic lung. Immunohistochemical analysis showed reduced expression of VEGFB protein in hypoxia although PlGF protein was unchanged. The expression of VEGFA mRNA and protein was unchanged. In vitro PlGF at high concentration mimicked the wound-healing actions of VEGFA on pulmonary microvascular endothelial monolayers. Low concentrations of PlGF potentiated the wound-healing actions of VEGFA while higher concentrations of PlGF were without this effect. VEGFB inhibited the wound-healing actions of VEGFA while VEGFB and PlGF together were mutually antagonistic. CONCLUSIONS VEGFB and PlGF can either inhibit or potentiate the actions of VEGFA, depending on their relative concentrations, which change in the hypoxic lung. Thus their actions in vivo depend on their specific concentrations within the microenvironment of the alveolar wall during the course of adaptation to pulmonary hypoxia.
Collapse
Affiliation(s)
- Michelle Sands
- School of Medicine and Medical Science, Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin, Ireland
| | | | | | | |
Collapse
|
95
|
Vanderpool RR, Kim AR, Molthen R, Chesler NC. Effects of acute Rho kinase inhibition on chronic hypoxia-induced changes in proximal and distal pulmonary arterial structure and function. J Appl Physiol (1985) 2011; 110:188-98. [PMID: 21088209 PMCID: PMC3253002 DOI: 10.1152/japplphysiol.00533.2010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Accepted: 11/11/2010] [Indexed: 01/25/2023] Open
Abstract
Hypoxic pulmonary hypertension (HPH) is initially a disease of the small pulmonary arteries. Its severity is usually quantified by pulmonary vascular resistance (PVR). Acute Rho kinase inhibition has been found to reduce PVR toward control values in animal models, suggesting that persistent pulmonary vasoconstriction is the dominant mechanism for increased PVR. However, HPH may also cause proximal arterial changes, which are relevant to right ventricular (RV) afterload. RV afterload can be quantified by pulmonary vascular impedance, which is obtained via spectral analysis of pulsatile pressure-flow relationships. To determine the effects of HPH independent of persistent pulmonary vasoconstriction in proximal and distal arteries, we quantified pulsatile pressure-flow relationships before and after acute Rho kinase inhibition and measured pulmonary arterial structure with microcomputed tomography. In control lungs, Rho kinase inhibition decreased 0 Hz impedance (Z₀), which is equivalent to PVR, from 2.1 ± 0.4 to 1.5 ± 0.2 mmHg·min·ml⁻¹ (P < 0.05) and tended to increase characteristic impedance (Z(C)) from 0.21 ± 0.01 to 0.22 ± 0.01 mmHg·min·ml⁻¹. In HPH lungs, Rho kinase inhibition decreased Z₀ (P < 0.05) without affecting Z(C). Microcomputed tomography measurements performed on lungs after acute Rho kinase inhibition demonstrated that HPH significantly decreased the unstressed diameter of the main pulmonary artery (760 ± 60 vs. 650 ± 80 μm; P < 0.05), decreased right pulmonary artery compliance, and reduced the frequency of arteries of diameter 50-100 μm (both P < 0.05). These results demonstrate that acute Rho kinase inhibition reverses many but not all HPH-induced changes in distal pulmonary arteries but does not affect HPH-induced changes in the conduit arteries that impact RV afterload.
Collapse
Affiliation(s)
- Rebecca R Vanderpool
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, USA
| | | | | | | |
Collapse
|
96
|
Nunes KP, Rigsby CS, Webb RC. RhoA/Rho-kinase and vascular diseases: what is the link? Cell Mol Life Sci 2010; 67:3823-36. [PMID: 20668910 PMCID: PMC2996825 DOI: 10.1007/s00018-010-0460-1] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Revised: 07/07/2010] [Accepted: 07/08/2010] [Indexed: 12/29/2022]
Abstract
RhoA/Rho-kinase pathway plays an important role in many pathological conditions. RhoA participates in the regulation of smooth muscle tone and activates many downstream kinases. The best characterized are the serine/threonine kinase isoforms (Rho-kinase or ROCK), ROCKα/ROCK2 and ROCKβ/ROCK1. ROCK is necessary for diverse functions such as local blood flow, arterial/pulmonary blood pressure, airway resistance and intestinal peristalsis. ROCK activation permits actin/myosin interactions and smooth muscle cells contraction by maintaining the activity of myosin light-chain kinase, independently of the free cytosolic calcium level. The sensitization of smooth muscle myofilaments to calcium has been implicated in many pathological states, such as hypertension, diabetes, heart attack, stroke, pulmonary hypertension, erectile dysfunction, and cancer. The focus of this review is on the involvement of RhoA/Rho-kinase in diseases. We will briefly describe the ROCK isoforms and the role of RhoA/Rho-kinase in the vasculature, before exploring the most recent findings regarding this pathway and various diseases.
Collapse
Affiliation(s)
- Kenia Pedrosa Nunes
- Department of Physiology, Medical College of Georgia, Augusta, GA 30912, USA.
| | | | | |
Collapse
|
97
|
Yamaji-Kegan K, Su Q, Angelini DJ, Myers AC, Cheadle C, Johns RA. Hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELMalpha) increases lung inflammation and activates pulmonary microvascular endothelial cells via an IL-4-dependent mechanism. THE JOURNAL OF IMMUNOLOGY 2010; 185:5539-48. [PMID: 20889544 DOI: 10.4049/jimmunol.0904021] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Hypoxia-induced mitogenic factor (HIMF), also known as found in inflammatory zone 1 and resistin-like molecule α, belongs to a novel class of cysteine-rich secreted proteins. It exhibits mitogenic and chemotactic properties during pulmonary hypertension-associated vascular remodeling, as well as fibrogenic properties during pulmonary fibrosis. HIMF expression in the lung was reported to be regulated by Th2 cytokines (IL-4 and IL-13) via the transcription factor STAT6 pathway in a bleomycin-induced pulmonary fibrosis model. However, in this study, we found that in the hypoxia-induced pulmonary hypertension model, lung HIMF expression is increased in IL-4 and STAT6 knockout (KO) mice to the same degree as in wild-type (WT) mice, suggesting that induction of HIMF expression does not require Th2 regulation in this model. We also found that HIMF-induced proliferative activity, hypertrophy, collagen, and extracellular matrix deposition in the pulmonary arteries are significantly less in IL-4 KO mice than in WT mice. In addition, HIMF-induced production of angiogenic factors/chemokines, such as vascular endothelial growth factor, MCP-1, and stromal-derived factor-1, in the lung resident cells, as well as macrophage infiltration, were significantly suppressed in the lungs of IL-4 KO mice. We also show that IL-4 was significantly increased in the lungs of HIMF-treated WT mice. Our in vitro studies using pulmonary microvascular endothelial cells revealed that HIMF stimulated cell proliferation, vascular endothelial growth factor expression, and MCP-1 production in a manner that is dependent on the IL-4/IL-4Rα system. These findings suggest that IL-4 signaling may play a significant role in HIMF-induced lung inflammation and vascular remodeling.
Collapse
Affiliation(s)
- Kazuyo Yamaji-Kegan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|
98
|
Abstract
During the development of the pulmonary vasculature in the fetus, many structural and functional changes occur to prepare the lung for the transition to air breathing. The development of the pulmonary circulation is genetically controlled by an array of mitogenic factors in a temporo-spatial order. With advancing gestation, pulmonary vessels acquire increased vasoreactivity. The fetal pulmonary vasculature is exposed to a low oxygen tension environment that promotes high intrinsic myogenic tone and high vasocontractility. At birth, a dramatic reduction in pulmonary arterial pressure and resistance occurs with an increase in oxygen tension and blood flow. The striking hemodynamic differences in the pulmonary circulation of the fetus and newborn are regulated by various factors and vasoactive agents. Among them, nitric oxide, endothelin-1, and prostaglandin I2 are mainly derived from endothelial cells and exert their effects via cGMP, cAMP, and Rho kinase signaling pathways. Alterations in these signaling pathways may lead to vascular remodeling, high vasocontractility, and persistent pulmonary hypertension of the newborn.
Collapse
Affiliation(s)
- Yuansheng Gao
- Department of Physiology and Pathophysiology, Peking University, Health Science Center, Beijing, China; and Department of Pediatrics, University of Illinois, College of Medicine at Chicago, Chicago, Illinois
| | - J. Usha Raj
- Department of Physiology and Pathophysiology, Peking University, Health Science Center, Beijing, China; and Department of Pediatrics, University of Illinois, College of Medicine at Chicago, Chicago, Illinois
| |
Collapse
|
99
|
Connolly MJ, Aaronson PI. Key role of the RhoA/Rho kinase system in pulmonary hypertension. Pulm Pharmacol Ther 2010; 24:1-14. [PMID: 20833255 DOI: 10.1016/j.pupt.2010.09.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 07/06/2010] [Accepted: 09/02/2010] [Indexed: 02/06/2023]
Abstract
Pulmonary hypertension (PH) is a general term comprising a spectrum of pulmonary hypertensive disorders which have in common an elevation of mean pulmonary arterial pressure (mPAP). The prototypical form of the disease, termed pulmonary arterial hypertension (PAH), is a rare but lethal syndrome with a complex aetiology characterised by increased pulmonary vascular resistance (PVR) and progressive elevation of mPAP; patients generally die from heart failure. Current therapies are inadequate and median survival is less than three years. PH due to chronic hypoxia (CH) is a condition separate from PAH and is strongly associated with chronic obstructive pulmonary disease (COPD). An early event in the pathogenesis of this form of PH is hypoxic pulmonary vasoconstriction (HPV), an acute homeostatic process that maintains the ventilation-perfusion ratio during alveolar hypoxia. The mechanisms underlying HPV remain controversial, but RhoA/Rho kinase (ROK)-mediated Ca²+-sensitisation is considered important. Increasing evidence also implicates RhoA/ROK in PASMC proliferation, inflammatory cell recruitment and the regulation of cell motility, all of which are involved in the pulmonary vascular remodelling occurring in all forms of PH. ROK is therefore a potential therapeutic target in treating PH of various aetiologies. Here, we examine current concepts regarding the aetiology of PAH and also PH due to CH, focusing on the contribution that RhoA/ROK-mediated processes may make to their development and on ROK inhibitors as potential therapies.
Collapse
Affiliation(s)
- Michelle J Connolly
- Division of Asthma, Allergy and Lung Biology, School of Medicine, King's College London, United Kingdom.
| | | |
Collapse
|
100
|
Casey DB, Badejo AM, Dhaliwal JS, Sikora JL, Fokin A, Golwala NH, Greco AJ, Murthy SN, Nossaman BD, Hyman AL, Kadowitz PJ. Analysis of responses to the Rho-kinase inhibitor Y-27632 in the pulmonary and systemic vascular bed of the rat. Am J Physiol Heart Circ Physiol 2010; 299:H184-92. [PMID: 20435851 PMCID: PMC2904133 DOI: 10.1152/ajpheart.00181.2009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Accepted: 04/26/2010] [Indexed: 12/21/2022]
Abstract
Responses to the Rho kinase inhibitor Y-27632 were investigated in the anesthetized rat. Under baseline conditions intravenous injections of Y-27632 decreased pulmonary and systemic arterial pressures and increased cardiac output. The decreases in pulmonary arterial pressures were enhanced when baseline tone was increased with U-46619, and under elevated tone conditions Y-27632 produced similar percent decreases in pulmonary and systemic arterial pressures. Injections of Y-27632 prevented and reversed the hypoxic pulmonary vasoconstrictor response. The increase in pulmonary arterial pressure in response to ventilation with a 10% O(2)-90% N(2) gas mixture was not well maintained during the period of hypoxic exposure. Treatment with the nitric oxide (NO) synthase (NOS) inhibitor nitro-l-arginine methyl ester (l-NAME) increased pulmonary arterial pressure and prevented the decline or fade in the hypoxic pulmonary vasoconstrictor response. The hypoxic pulmonary vasoconstrictor response was reversed by Y-27632 in control and in l-NAME-treated animals. The Rho kinase inhibitor attenuated increases in pulmonary arterial pressures in response to intravenous injections of serotonin, angiotensin II, and Bay K 8644. Y-27632, sodium nitrite, and BAY 41-8543, a guanylate cyclase stimulator, decreased pulmonary and systemic arterial pressures and vascular resistances in monocrotaline-treated rats. These data suggest that Rho kinase is involved in the regulation of baseline tone and in the mediation of pulmonary vasoconstrictor responses. The present data suggest that the hypoxic pulmonary vasoconstrictor response is modulated by the release of NO that mediates the nonsustained component of the response in the anesthetized rat. These data suggest that Rho kinase and NOS play important roles in the regulation of vasoconstrictor tone in physiological and pathophysiological states and that monocrotaline-induced pulmonary hypertension can be reversed by agents that inhibit Rho kinase, generate NO, or stimulate soluble guanylate cyclase.
Collapse
Affiliation(s)
- David B Casey
- Department of Pharmacology, Health Sciences Center, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|