1
|
Olave NC, Halloran B, Ambalavanan N. FGF2 is secreted in extracellular vesicles from lung cells. Am J Physiol Lung Cell Mol Physiol 2024; 327:L359-L370. [PMID: 39010825 PMCID: PMC11444508 DOI: 10.1152/ajplung.00225.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 06/11/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
The 18-kDa isoform of basic fibroblast growth factor (bFGF/FGF2) lacks a conventional signal peptide sequence and is exported by a novel membrane-associated transport pathway. Extracellular vesicles (EVs) are increasingly recognized as mediators of intercellular communication in the lung, and our prior work demonstrates that EVs carry cargo that contributes to hyperoxic lung injury and are biomarkers for bronchopulmonary dysplasia. We used primary human bronchial epithelial (HBE), pulmonary artery endothelial (HPAE), and fibroblast (HNF) cells to determine whether FGF2 was secreted in EVs. EVs were isolated by ultracentrifugation from HBE, HPAE, and HNF exposed to either normoxia or hyperoxia, followed by nanoparticle tracking analysis and electron microscopy. Hyperoxia exposure increased the total EV number. All three cell types released FGF2-18kDa both directly into the extracellular environment (secretome), as well as in EVs. HBE released more FGF2-18kDa in EVs during hyperoxia, and these were internalized and localized to both nuclei and cytoplasm of recipient cells. By co-immunoprecipitation, we identified potential binding partners of FGF2-18kDa in the nuclei, including histone 1.2 (H1.2) binding protein, that may mediate downstream effects that do not involve FGF2 binding to cell surface receptors. FGF2-18kDa interaction with H1.2 binding protein may indicate a mechanism by which FGF2 secreted in EVs modulates cellular processes. FGF2 was also found to increase angiogenesis by Matrigel assay. Further studies are necessary to determine the biological relevance of FGF2 in EVs as modulators of lung injury and disease.NEW & NOTEWORTHY We found that multiple lung cell types release basic fibroblast growth factor (FGF2)-18kDa both directly into the extracellular environment (secretome), as well as in extracellular vesicles (EVs). Bronchial epithelial cells released more FGF2-18kDa in EVs during hyperoxia, which could be internalized rapidly by recipient cells. We also identified potential binding partners of FGF2-18kDa in nuclei that may mediate downstream effects that do not involve FGF2 binding to cell surface receptors. We also confirmed a potential angiogenic role for FGF2-18kDa.
Collapse
Affiliation(s)
- Nelida C Olave
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Brian Halloran
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Namasivayam Ambalavanan
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
2
|
Mukherjee D, Konduri GG. Pediatric Pulmonary Hypertension: Definitions, Mechanisms, Diagnosis, and Treatment. Compr Physiol 2021; 11:2135-2190. [PMID: 34190343 PMCID: PMC8289457 DOI: 10.1002/cphy.c200023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pediatric pulmonary hypertension (PPH) is a multifactorial disease with diverse etiologies and presenting features. Pulmonary hypertension (PH), defined as elevated pulmonary artery pressure, is the presenting feature for several pulmonary vascular diseases. It is often a hidden component of other lung diseases, such as cystic fibrosis and bronchopulmonary dysplasia. Alterations in lung development and genetic conditions are an important contributor to pediatric pulmonary hypertensive disease, which is a distinct entity from adult PH. Many of the causes of pediatric PH have prenatal onset with altered lung development due to maternal and fetal conditions. Since lung growth is altered in several conditions that lead to PPH, therapy for PPH includes both pulmonary vasodilators and strategies to restore lung growth. These strategies include optimal alveolar recruitment, maintaining physiologic blood gas tension, nutritional support, and addressing contributing factors, such as airway disease and gastroesophageal reflux. The outcome for infants and children with PH is highly variable and largely dependent on the underlying cause. The best outcomes are for neonates with persistent pulmonary hypertension (PPHN) and reversible lung diseases, while some genetic conditions such as alveolar capillary dysplasia are lethal. © 2021 American Physiological Society. Compr Physiol 11:2135-2190, 2021.
Collapse
Affiliation(s)
- Devashis Mukherjee
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, 53226 USA
| | - Girija G. Konduri
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, 53226 USA
| |
Collapse
|
3
|
Outcomes of infants and children undergoing surgical repair of ventricular septal defect: a review of the literature and implications for research with an emphasis on pulmonary artery hypertension. Cardiol Young 2020; 30:799-806. [PMID: 32431266 DOI: 10.1017/s1047951120001146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Pulmonary vascular disease resulting from CHDs may be the most preventable cause of pulmonary artery hypertension worldwide. Many children in developing countries still do not have access to early closure of clinically significant defects, and the long-term outcomes after corrective surgery remain unclear. Focused on long-term results after isolated ventricular septal defect repair, our review sought to determine the most effective medical therapy for the pre-operative management of elevated left-to-right shunts in patients with an isolated ventricular septal defect. METHODS We identified articles specific to the surgical repair of isolated ventricular septal defects. Specific parameters included the pathophysiology and pre-operative medical management of pulmonary over-circulation and outcomes. RESULTS Studies most commonly focused on histologic changes to the pulmonary vasculature and levels of thromboxanes, prostaglandins, nitric oxide, endothelin, and matrix metalloproteinases. Only 2/44 studies mentioned targeted pharmacologic management to any of these systems related to ventricular septal defect repair; no study offered evidence-based guidelines to manage pulmonary over-circulation with ventricular septal defects. Most studies with long-term data indicated a measurable frequency of pulmonary artery hypertension or diminished exercise capacity late after ventricular septal defect repair. CONCLUSION Long-term pulmonary vascular and respiratory changes can occur in children after ventricular septal defect repair. Research should be directed at providing an evidenced-based approach to the medical management of infants and children with ventricular septal defects (and naturally all CHDs) to minimise consequences of pulmonary artery hypertension, particularly as defect repair may occur late in underprivileged societies.
Collapse
|
4
|
Ambade AS, Jung B, Lee D, Doods H, Wu D. Triple-tyrosine kinase inhibition attenuates pulmonary arterial hypertension and neointimal formation. Transl Res 2019; 203:15-30. [PMID: 30142307 DOI: 10.1016/j.trsl.2018.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/21/2018] [Accepted: 07/23/2018] [Indexed: 12/29/2022]
Abstract
The present study examined the effects of simultaneous inhibition of vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF) receptor signaling with BIBF1000, a novel triple tyrosine kinase inhibitor on preventing and reversing the progression of severe pulmonary arterial hypertension (PAH) in an experimental model in rats. Left pneumonectomized male Wistar rats were injected with monocrotaline to induce PAH. Treatment with BIBF1000 from day 1 to day 21 after monocrotaline injection attenuated PAH development, as evidenced by lower values for pulmonary artery pressure (mPAP), right ventricular pressure (RVSP), pulmonary arterial neointimal formation, and the ratio of right ventricular weight to left ventricular and septum weight [RV/(LV+S)] on day 21 compared to control rats. Treatment with BIBF1000 from day 21 to day 42 after monocrotaline injection reversed established PAH as shown by normalized values for mPAP and RVSP, RV/(LV+S) ratio, pulmonary arterial occlusion scores, levels of heart and lung fibrosis, as well as improved survival. Treatment with BIBF1000 reduced inflammatory cell recruitment in bronchoalveolar lavage and lung tissues, reduced CD-68 positive macrophages and expression of proliferating cell nuclear antigen in the perivascular areas, and reduced TNF-α and growth factor productions, and inhibited the phosphorylation of AKT and GSK3β in lungs. In addition, BIBF1000 inhibited pulmonary artery smooth muscle cells migration and proliferation from rat pulmonary artery explant cultures. Simultaneous inhibition of VEGF, PDGF, and FGF receptor signaling by BIBF1000 prevents and reverses the progression of severe pulmonary arterial hypertension and vascular remodeling in this experimental model.
Collapse
Affiliation(s)
- Anjira S Ambade
- Department of BIN Convergence Technology, Chonbuk National University, Jeonju, South Korea
| | - Birgit Jung
- Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Dongwon Lee
- Department of BIN Convergence Technology, Chonbuk National University, Jeonju, South Korea
| | - Henri Doods
- Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Dongmei Wu
- Department of BIN Convergence Technology, Chonbuk National University, Jeonju, South Korea; Department of Research, Mount Sinai Medical Center, Miami Beach, Florida.
| |
Collapse
|
5
|
Redox Mechanisms Influencing cGMP Signaling in Pulmonary Vascular Physiology and Pathophysiology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:227-240. [PMID: 29047089 DOI: 10.1007/978-3-319-63245-2_13] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The soluble form of guanylate cyclase (sGC) and cGMP signaling are major regulators of pulmonary vasodilation and vascular remodeling that protect the pulmonary circulation from hypertension development. Nitric oxide, reactive oxygen species, thiol and heme redox, and heme biosynthesis control mechanisms regulating the production of cGMP by sGC. In addition, a cGMP-independent mechanism regulates protein kinase G through thiol oxidation in manner controlled by peroxide metabolism and NADPH redox. Multiple aspects of these regulatory processes contribute to physiological and pathophysiological regulation of the pulmonary circulation, and create potentially novel therapeutic targets for the treatment of pulmonary vascular disease.
Collapse
|
6
|
Ketabchi F, Bajoovand S, Adlband M, Naseh M, Nekooeian AA, Mashghoolozekr E. Right ventricular pressure elevated in one-kidney, one clip Goldblatt hypertensive rats. Clin Exp Hypertens 2017; 39:344-349. [PMID: 28513232 DOI: 10.1080/10641963.2016.1259329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Both renal and respiratory diseases are common with high mortality rate around the world. This study was the first to compare effects of two kidneys, one clip (2K1C) and one-kidney, one clip (1K1C) Goldblatt hypertension on right ventricular pressure during normal condition and mechanical ventilation with hypoxia gas. Male Sprague-Dawley rats were subjected to control, 2K1C, or 1K1C groups. Twenty-eight days after the first surgery, animals were anesthetized, and femoral artery and vein, and right ventricle cannulated. Systemic arterial pressure and right ventricular systolic pressures (RVSP) were recorded during ventilation the animals with normoxic or hypoxic gas. RVSP in the 1K1C group was significantly more than the control and 2K1C groups during baseline conditions and ventilation the animals with hypoxic gas. Administration of antioxidant Trolox increased RVSP in the 1K1C and control groups compared with their baselines. Furthermore, there was no alteration in RVSP during hypoxia in the presence of Trolox. This study indicated that RVSP only increased after 28 days induction of 1K1C but not 2K1C model. In addition, it seems that the response to hypoxic gas and antioxidants in 1K1C is more than 2K1C. These data also suggest that effects of 1K1C may partially be related to reactive oxygen species (ROS) pathways.
Collapse
Affiliation(s)
- Farzaneh Ketabchi
- a Department of Physiology, School of Medicine , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Shirin Bajoovand
- b Department of Food and Drug, Reference Laboratory, School of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Mojtaba Adlband
- a Department of Physiology, School of Medicine , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Maryam Naseh
- a Department of Physiology, School of Medicine , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Ali A Nekooeian
- c Department of Phamacology, School of Medicine , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Elaheh Mashghoolozekr
- c Department of Phamacology, School of Medicine , Shiraz University of Medical Sciences , Shiraz , Iran
| |
Collapse
|
7
|
Idiopathic pre-capillary pulmonary hypertension in patients with end-stage kidney disease: effect of endothelin receptor antagonists. Clin Exp Nephrol 2016; 21:1088-1096. [PMID: 27757709 PMCID: PMC5698371 DOI: 10.1007/s10157-016-1344-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/06/2016] [Indexed: 11/16/2022]
Abstract
Background We examined the prevalence, prognosis, and effect of endothelin receptor antagonists on survival in end-stage kidney disease patients with idiopathic pre-capillary pulmonary hypertension. Methods We investigated 1988 end-stage kidney disease patients in Toujinkai Hospital from January 1, 2001 to December 31, 2014. Pulmonary hypertension was screened by symptoms (dyspnea, hypotension, or near syncope) and echocardiography, and diagnosed by computed tomography with enhancement, pulmonary flow scintigraphy, and right heart catheterization. Results Fifteen patients (67 ± 11 years; 12 women and 3 men) were diagnosed as idiopathic pre-capillary pulmonary hypertension; mean pulmonary arterial pressure, pulmonary vascular resistance, or pulmonary artery wedge pressure were 55 ± 11 mmHg, 7.5 ± 2.9 Woods units, or 12 ± 2 mmHg, respectively. Of the 15 patients, 14 received hemodialysis, and 1 was in a pre-dialysis stage. Patients were followed through December 31, 2015, and 11 died of heart failure; their mean survival time was 26.4 ± 21.0 months. Endothelin receptor antagonists were used for 11 patients, and mean survival times were 57.3 ± 12.1 months in patients with endothelin receptor antagonists and 7.5 ± 2.1 months in those without. In the Kaplan–Meier analysis, heart failure death-free survival rates were higher in patients with endothelin receptor antagonists than in those without (P < 0.001); 100 versus 25 % at one year and 71 versus 0 % at 3 years. Conclusion The prognosis of idiopathic pre-capillary pulmonary hypertension seems to be poor in end-stage kidney disease patients. Administration of endothelin receptor antagonists might improve the survival by inhibiting heart failure death. Registration of clinical trials This study was registered to the ClinicalTrials.gov (https://clinicaltrials.gov/): protocol identifier, NCT02743091.
Collapse
|
8
|
Zheng Y, Ma H, Hu E, Huang Z, Cheng X, Xiong C. Inhibition of FGFR Signaling With PD173074 Ameliorates Monocrotaline-induced Pulmonary Arterial Hypertension and Rescues BMPR-II Expression. J Cardiovasc Pharmacol 2015; 66:504-14. [DOI: 10.1097/fjc.0000000000000302] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
9
|
Zhang YF, Zheng Y. The effects of mycophenolate mofetil on cytokines and their receptors in pulmonary arterial hypertension in rats. Scand J Rheumatol 2015; 44:412-5. [DOI: 10.3109/03009742.2015.1023829] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
10
|
Chang YT, Tseng CN, Tannenberg P, Eriksson L, Yuan K, de Jesus Perez VA, Lundberg J, Lengquist M, Botusan IR, Catrina SB, Tran PK, Hedin U, Tran-Lundmark K. Perlecan heparan sulfate deficiency impairs pulmonary vascular development and attenuates hypoxic pulmonary hypertension. Cardiovasc Res 2015; 107:20-31. [PMID: 25952902 DOI: 10.1093/cvr/cvv143] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 05/01/2015] [Indexed: 12/21/2022] Open
Abstract
AIMS Excessive vascular cell proliferation is an important component of pulmonary hypertension (PH). Perlecan is the major heparan sulfate (HS) proteoglycan in the vascular extracellular matrix. It binds growth factors, including FGF2, and either restricts or promotes cell proliferation. In this study, we have explored the effects of perlecan HS deficiency on pulmonary vascular development and in hypoxia-induced PH. METHODS AND RESULTS In normoxia, Hspg2(Δ3/Δ3) mice, deficient in perlecan HS, had reduced pericytes and muscularization of intra-acinar vessels. Pulmonary angiography revealed a peripheral perfusion defect. Despite these abnormalities, right ventricular systolic pressure (RVSP) and myocardial mass remained normal. After 4 weeks of hypoxia, increases in the proportion of muscularized vessels, RVSP, and right ventricular hypertrophy were significantly less in Hspg2(Δ3/Δ3) compared with wild type. The early phase of hypoxia induced a significantly lower increase in fibroblast growth factor receptor-1 (FGFR1) protein level and receptor phosphorylation, and reduced pulmonary artery smooth muscle cell (PASMC) proliferation in Hspg2(Δ3/Δ3). At 4 weeks, FGF2 mRNA and protein were also significantly reduced in Hspg2(Δ3/Δ3) lungs. Ligand and carbohydrate engagement assay showed that perlecan HS is required for HS-FGF2-FGFR1 ternary complex formation. In vitro, proliferation assays showed that PASMC proliferation is reduced by selective FGFR1 inhibition. PASMC adhesion to fibronectin was higher in Hspg2(Δ3/Δ3) compared with wild type. CONCLUSIONS Perlecan HS chains are important for normal vascular arborization and recruitment of pericytes to pulmonary vessels. Perlecan HS deficiency also attenuates hypoxia-induced PH, where the underlying mechanisms involve impaired FGF2/FGFR1 interaction, inhibition of PASMC growth, and altered cell-matrix interactions.
Collapse
Affiliation(s)
- Ya-Ting Chang
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Chi-Nan Tseng
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Philip Tannenberg
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Linnéa Eriksson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ke Yuan
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Vinicio A de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Johan Lundberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Mariette Lengquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | | | - Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Phan-Kiet Tran
- Department of Cardiothoracic Surgery, Uppsala University, Uppsala, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Karin Tran-Lundmark
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
11
|
Shirai Y, Okazaki Y, Inoue Y, Tamura Y, Yasuoka H, Takeuchi T, Kuwana M. Elevated levels of pentraxin 3 in systemic sclerosis: associations with vascular manifestations and defective vasculogenesis. Arthritis Rheumatol 2015; 67:498-507. [PMID: 25385504 DOI: 10.1002/art.38953] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 11/06/2014] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To clarify the role of pentraxin 3 (PTX3), a multifunctional pattern recognition protein that can suppress fibroblast growth factor 2 (FGF-2), in systemic sclerosis (SSc)-related vasculopathy. METHODS We assessed 171 SSc patients and 19 age- and sex-matched healthy control subjects. Circulating PTX3 and FGF-2 levels were measured by enzyme immunoassay, and CD34+CD133+CD309+ endothelial progenitor cells (EPCs) were counted by flow cytometry. Correlations between PTX3 and FGF-2 and the presence or future development of vascular manifestations, including digital ulcers and pulmonary arterial hypertension (PAH), were identified by univariate and multivariate analysis. The effect of PTX3 on EPC differentiation was evaluated in proangiogenic cultures of mouse bone marrow cells in combination with colony formation assay. RESULTS Circulating PTX3 and FGF-2 levels were significantly higher in SSc patients than in healthy control subjects. PTX3 was elevated in SSc patients who had digital ulcers or PAH, while FGF-2 was reduced in SSc patients with PAH. Multivariate analysis identified elevated PTX3 as an independent parameter associated with the presence of digital ulcers and PAH, and PTX3 levels were a useful predictor of future occurrences of digital ulcers. Reduced FGF-2 was independently associated with the presence of PAH. EPC counts were significantly lower in patients with digital ulcers or PAH and correlated negatively with circulating PTX3 concentrations. Finally, PTX3 inhibited EPC differentiation in vitro. CONCLUSION In SSc patients, exposure to high concentrations of PTX3 may suppress EPC-mediated vasculogenesis and promote vascular manifestations such as digital ulcers and PAH.
Collapse
|
12
|
Wedgwood S, Steinhorn RH. Role of reactive oxygen species in neonatal pulmonary vascular disease. Antioxid Redox Signal 2014; 21:1926-42. [PMID: 24350610 PMCID: PMC4202910 DOI: 10.1089/ars.2013.5785] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Abnormal lung development in the perinatal period can result in severe neonatal complications, including persistent pulmonary hypertension (PH) of the newborn and bronchopulmonary dysplasia. Reactive oxygen species (ROS) play a substantive role in the development of PH associated with these diseases. ROS impair the normal pulmonary artery (PA) relaxation in response to vasodilators, and ROS are also implicated in pulmonary arterial remodeling, both of which can increase the severity of PH. RECENT ADVANCES PA ROS levels are elevated when endogenous ROS-generating enzymes are activated and/or when endogenous ROS scavengers are inactivated. Animal models have provided valuable insights into ROS generators and scavengers that are dysregulated in different forms of neonatal PH, thus identifying potential therapeutic targets. CRITICAL ISSUES General antioxidant therapy has proved ineffective in reversing PH, suggesting that it is necessary to target specific signaling pathways for successful therapy. FUTURE DIRECTIONS Development of novel selective pharmacologic inhibitors along with nonantioxidant therapies may improve the treatment outcomes of patients with PH, while further investigation of the underlying mechanisms may enable earlier detection of the disease.
Collapse
Affiliation(s)
- Stephen Wedgwood
- Department of Pediatrics, University of California Davis Medical Center , Sacramento, California
| | | |
Collapse
|
13
|
Datar SA, Oishi PE, Gong W, Bennett SH, Sun CE, Johengen M, Maki J, Johnson RC, Raff GW, Fineman JR. Altered reactivity and nitric oxide signaling in the isolated thoracic duct from an ovine model of congenital heart disease with increased pulmonary blood flow. Am J Physiol Heart Circ Physiol 2014; 306:H954-62. [PMID: 24531811 DOI: 10.1152/ajpheart.00841.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
We have previously shown decreased pulmonary lymph flow in our lamb model of chronically increased pulmonary blood flow, created by the in utero placement of an 8-mm aortopulmonary shunt. The purpose of this study was to test the hypothesis that abnormal lymphatic function in shunt lambs is due to impaired lymphatic endothelial nitric oxide (NO)-cGMP signaling resulting in increased lymphatic vascular constriction and/or impaired relaxation. Thoracic duct rings were isolated from 4-wk-old shunt (n = 7) and normal (n = 7) lambs to determine length-tension properties, vascular reactivity, and endothelial NO synthase protein. At baseline, shunt thoracic duct rings had 2.6-fold higher peak to peak tension and a 2-fold increase in the strength of contractions compared with normal rings (P < 0.05). In response to norepinephrine, shunt thoracic duct rings had a 2.4-fold increase in vascular tone compared with normal rings (P < 0.05) and impaired relaxation in response to the endothelium-dependent dilator acetylcholine (63% vs. 13%, P < 0.05). In vivo, inhaled NO (40 ppm) increased pulmonary lymph flow (normalized for resistance) ∼1.5-fold in both normal and shunt lambs (P < 0.05). Inhaled NO exposure increased bioavailable NO [nitrite/nitrate (NOx); ∼2.5-fold in normal lambs and ∼3.4-fold in shunt lambs] and cGMP (∼2.5-fold in both) in the pulmonary lymph effluent (P < 0.05). Chronic exposure to increased pulmonary blood flow is associated with pulmonary lymphatic endothelial injury that disrupts NO-cGMP signaling, leading to increased resting vasoconstriction, increased maximal strength of contraction, and impaired endothelium-dependent relaxation. Inhaled NO increases pulmonary lymph NOx and cGMP levels and pulmonary lymph flow in normal and shunt lambs. Therapies that augment NO-cGMP signaling within the lymphatic system may provide benefits, warranting further study.
Collapse
Affiliation(s)
- Sanjeev A Datar
- Department of Pediatrics, University of California, San Francisco, California
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
El Chami H, Hassoun PM. Inflammatory mechanisms in the pathogenesis of pulmonary arterial hypertension. Compr Physiol 2013; 1:1929-41. [PMID: 23733693 DOI: 10.1002/cphy.c100028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inflammation is a prominent feature of human and experimental pulmonary hypertension (PH) as suggested by infiltration of various inflammatory cells and increased expression of certain cytokines in remodeled pulmonary vessels. Macrophages, T and B lymphocytes, and dendritic cells are found in the vascular lesions of idiopathic pulmonary arterial hypertension (PAH) as well as in PAH associated with connective tissue diseases or infectious etiologies such as HIV. In addition, PAH is often characterized by the presence of circulating chemokines and cytokines, increased expression of growth (such as VEGF and PDGF) and transcriptional (e.g., nuclear factor of activated T cells or NFAT) factors, and viral protein components (e.g., HIV-1 Nef), which directly contribute to further recruitment of inflammatory cells and the pulmonary vascular remodeling process. These inflammatory pathways may thus serve as potential specific therapeutic targets. This article provides an overview of inflammatory pathways involving chemokines and cytokines as well as growth factors, highlighting their potential role in pulmonary vascular remodeling and the possibility of future targeted therapy.
Collapse
Affiliation(s)
- Hala El Chami
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | | |
Collapse
|
15
|
Aggarwal S, Gross CM, Sharma S, Fineman JR, Black SM. Reactive oxygen species in pulmonary vascular remodeling. Compr Physiol 2013; 3:1011-34. [PMID: 23897679 DOI: 10.1002/cphy.c120024] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The pathogenesis of pulmonary hypertension is a complex multifactorial process that involves the remodeling of pulmonary arteries. This remodeling process encompasses concentric medial thickening of small arterioles, neomuscularization of previously nonmuscular capillary-like vessels, and structural wall changes in larger pulmonary arteries. The pulmonary arterial muscularization is characterized by vascular smooth muscle cell hyperplasia and hypertrophy. In addition, in uncontrolled pulmonary hypertension, the clonal expansion of apoptosis-resistant endothelial cells leads to the formation of plexiform lesions. Based upon a large number of studies in animal models, the three major stimuli that drive the vascular remodeling process are inflammation, shear stress, and hypoxia. Although, the precise mechanisms by which these stimuli impair pulmonary vascular function and structure are unknown, reactive oxygen species (ROS)-mediated oxidative damage appears to play an important role. ROS are highly reactive due to their unpaired valence shell electron. Oxidative damage occurs when the production of ROS exceeds the quenching capacity of the antioxidant mechanisms of the cell. ROS can be produced from complexes in the cell membrane (nicotinamide adenine dinucleotide phosphate-oxidase), cellular organelles (peroxisomes and mitochondria), and in the cytoplasm (xanthine oxidase). Furthermore, low levels of tetrahydrobiopterin (BH4) and L-arginine the rate limiting cofactor and substrate for endothelial nitric oxide synthase (eNOS), can cause the uncoupling of eNOS, resulting in decreased NO production and increased ROS production. This review will focus on the ROS generation systems, scavenger antioxidants, and oxidative stress associated alterations in vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Saurabh Aggarwal
- Pulmonary Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta, Georgia, USA
| | | | | | | | | |
Collapse
|
16
|
Di Lullo L, Floccari F, Rivera R, Barbera V, Granata A, Otranto G, Mudoni A, Malaguti M, Santoboni A, Ronco C. Pulmonary Hypertension and Right Heart Failure in Chronic Kidney Disease: New Challenge for 21st-Century Cardionephrologists. Cardiorenal Med 2013; 3:96-103. [PMID: 23922549 DOI: 10.1159/000350952] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Pulmonary hypertension is defined as an increased systolic pulmonary pressure of >30 mm Hg, and it shows a 40% prevalence in hemodialysis patients due to vascular access (both central venous catheter and arteriovenous fistula). Secondary pulmonary hypertension in chronic kidney disease patients is strictly related to pulmonary circulation impairment together with chronic volume overload and increased levels of cytokines and growth factors, such as FGF, PDGF, and TGF-β, leading to fibrosis. Endothelial dysfunction, together with lower activation of NOS, increased levels of serum endothelin and fibrin storages, involves an extensive growth of endothelial cells leading to complete obliteration of pulmonary vessels. Pulmonary hypertension has no pathognomonic and distinctive symptoms and signs; standard transthoracic echocardiography allows easy assessment of compliance of the right heart chambers. The therapeutic approach is based on traditional drugs such as digitalis-derived drugs, vasodilatory agents (calcium channel blockers), and oral anticoagulants. New pharmacological agents are under investigation, such as prostaglandin analogues, endothelin receptor blockers, and phosphodiesterase-5 inhibitors.
Collapse
Affiliation(s)
- L Di Lullo
- Department of Nephrology and Dialysis, L. Parodi Delfino Hospital, Colleferro, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Altered immunity and inflammation are increasingly recognized features of pulmonary arterial hypertension (PAH). This is suggested by infiltration of various inflammatory cells (e.g., macrophages, T and B lymphocytes), increased cytokine and growth factor (e.g., VEGF and PDGF) expression in remodeled pulmonary vessels, and the presence of circulating chemokines and cytokines. In certain diseases associated with PAH, increased expression of growth and transcriptional (e.g., nuclear factor of activated T cells or NFAT) factors, and viral protein components (e.g., HIV-1 Nef), appear to contribute directly to recruitment of inflammatory cells in remodeled vessels, and may potentially serve as specific therapeutic targets. This section provides an overview of inflammatory pathways highlighting their potential role in pulmonary vascular remodeling in PAH and the possibility of future targeted therapy.
Collapse
Affiliation(s)
- Hala El Chami
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | | |
Collapse
|
18
|
Kojonazarov B, Sydykov A, Pullamsetti SS, Luitel H, Dahal BK, Kosanovic D, Tian X, Majewski M, Baumann C, Evans S, Phillips P, Fairman D, Davie N, Wayman C, Kilty I, Weissmann N, Grimminger F, Seeger W, Ghofrani HA, Schermuly RT. Effects of multikinase inhibitors on pressure overload-induced right ventricular remodeling. Int J Cardiol 2012; 167:2630-7. [PMID: 22854298 DOI: 10.1016/j.ijcard.2012.06.129] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 04/16/2012] [Accepted: 06/24/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Little is known about the effects of current PAH therapies and receptor tyrosine kinase inhibitors on heart remodeling. We sought to investigate the effects of the multikinase inhibitors sunitinib (PDGFR-, VEGFR- and KIT-inhibitor) and sorafenib (raf1/b-, VEGFR-, PDGFR-inhibitor) on pressure overload induced right ventricular (RV) remodeling. METHODS We investigated the effects of the kinase inhibitors on hemodynamics and remodeling in rats subjected either to monocrotaline (MCT)-induced PH or to surgical pulmonary artery banding (PAB). MCT rats were treated from days 21 to 35 with either vehicle, sunitinib (1mg/kg, 5mg/kg and 10mg/kg/day) or sorafenib (10mg/kg/day). PAB rats were treated with vehicle, sunitinib (10mg/kg/day) or sorafenib (10mg/kg/day) from days 7 to 21. RV function and remodeling were determined using echocardiography, invasive hemodynamic measurement and histomorphometry. RESULTS Treatment with both sorafenib and sunitinib decreased right ventricular systolic pressure, pulmonary vascular remodeling, RV hypertrophy and fibrosis in MCT rats. This was associated with an improvement of RV function. Importantly, after PAB, both compounds reversed RV chamber and cellular hypertrophy, reduced RV interstitial and perivascular fibrosis, and improved RV function. CONCLUSION We demonstrated that sunitinib and sorafenib reversed RV remodeling and significantly improved RV function measured via a range of invasive and non-invasive cardiopulmonary endpoints in experimental models of RV hypertrophy.
Collapse
Affiliation(s)
- Baktybek Kojonazarov
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Aggarwal S, Gross C, Fineman JR, Black SM. Oxidative stress and the development of endothelial dysfunction in congenital heart disease with increased pulmonary blood flow: lessons from the neonatal lamb. Trends Cardiovasc Med 2012; 20:238-46. [PMID: 22293025 DOI: 10.1016/j.tcm.2011.11.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Congenital heart diseases associated with increased pulmonary blood flow commonly leads to the development of pulmonary hypertension. However, most patients who undergo histological evaluation have advanced pulmonary hypertension, and therefore it has been difficult to investigate aberrations in signaling cascades that precede the development of overt vascular remodeling. This review discusses the role played by both oxidative and nitrosative stress in the lung and their impact on the signaling pathways that regulate vasodilation, vessel growth, and vascular remodeling in the neonatal lung exposed to increased pulmonary blood flow.
Collapse
Affiliation(s)
- Saurabh Aggarwal
- Pulmonary Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | | | |
Collapse
|
20
|
Fibrosis of pulmonary vascular remodeling in carotid artery-jugular vein shunt pulmonary artery hypertension model of rats. Eur J Cardiothorac Surg 2012; 41:162-6. [PMID: 21893417 DOI: 10.1016/j.ejcts.2011.04.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE The aim of the present study was to observe the changes of hemodynamics, stereology in pulmonary vascular remodeling and messenger RNA (mRNA) expressions of transforming growth factor beta 1, and receptors in carotid artery-jugular vein (CA-JV) shunt pulmonary artery hypertension model of rats. METHODS Thirty-six Sprague-Dawley rats were randomized into three groups: CA-JV group, monocrotaline (MCT) administration group, and control group. Left CA-JV shunts were established in CA-JV group. Dorsal subcutaneous injections of MCT (60 mg kg(-1)) were received in MCT group. Ligations of left common carotid artery and external jugular vein were performed in control group. Right ventricular systolic pressure (RVSP) measurement, histological evaluation of the pulmonary tissue, and mRNA levels of transforming growth factor beta 1 (TGFß1), receptor 1 and receptor 2, were investigated after 6 weeks on MCT group, and after 12 weeks on both control and CA-JV groups. RESULTS Compared with control group, RVSP, percentage of fibrous tissue (F%) in pulmonary arterioles, mRNA levels of TGFß1, and receptors of CA-JVand MCT groups increased significantly. Severe hemodynamics change was found in MCT groups. On the other hand, CA-JV group demonstrated more obvious fibrogenesis and TGFß1 signals' upregulation in two pulmonary artery hypertension (PAH) models. CONCLUSIONS CA-JV shunt model of rats was a well-established PAH animal model simulating congenital heart disease with systemic-pulmonary shunt.
Collapse
|
21
|
Abstract
Our understanding of, and approach to, pulmonary arterial hypertension has undergone a paradigm shift in the past decade. Once a condition thought to be dominated by increased vasoconstrictor tone and thrombosis, pulmonary arterial hypertension is now seen as a vasculopathy in which structural changes driven by excessive vascular cell growth and inflammation, with recruitment and infiltration of circulating cells, play a major role. Perturbations of a number of molecular mechanisms have been described, including pathways involving growth factors, cytokines, metabolic signaling, elastases, and proteases, that may underlie the pathogenesis of the disease. Elucidating their contribution to the pathophysiology of pulmonary arterial hypertension could offer new drug targets. The role of progenitor cells in vascular repair is also under active investigation. The right ventricular response to increased pressure load is recognized as critical to survival and the molecular mechanisms involved are attracting increasing interest. The challenge now is to integrate this new knowledge and explore how it can be used to categorize patients by molecular phenotype and tailor treatment more effectively.
Collapse
Affiliation(s)
- Ralph T. Schermuly
- Max-Planck-Institute for Heart and Lung Research, Parkstrasse 1, Bad Nauheim, 61231 Germany
| | - Hossein A. Ghofrani
- University Hospital Giessen and Marburg, University of Giessen Lung Center, Klinikstrasse 36, Giessen, 35392 Germany
| | - Martin R. Wilkins
- Division of Experimental Medicine, Centre for Pharmacology and Therapeutics, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN UK
| | - Friedrich Grimminger
- University Hospital Giessen and Marburg, University of Giessen Lung Center, Klinikstrasse 36, Giessen, 35392 Germany
| |
Collapse
|
22
|
Sklepkiewicz P, Schermuly RT, Tian X, Ghofrani HA, Weissmann N, Sedding D, Kashour T, Seeger W, Grimminger F, Pullamsetti SS. Glycogen synthase kinase 3beta contributes to proliferation of arterial smooth muscle cells in pulmonary hypertension. PLoS One 2011; 6:e18883. [PMID: 21533110 PMCID: PMC3078925 DOI: 10.1371/journal.pone.0018883] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 03/24/2011] [Indexed: 01/17/2023] Open
Abstract
Rationale Pulmonary arterial hypertension (PAH) is a rare progressive pulmonary vascular disorder associated with vascular remodeling and right heart failure. Vascular remodeling involves numerous signaling cascades governing pulmonary arterial smooth muscle cell (PASMC) proliferation, migration and differentiation. Glycogen synthase kinase 3beta (GSK3ß) is a serine/threonine kinase and can act as a downstream regulatory switch for numerous signaling pathways. Hence, we hypothesized that GSK3ß plays a crucial role in pulmonary vascular remodeling. Methods All experiments were done with lung tissue or isolated PASMCs in a well-established monocrotaline (MCT)-induced PAH rat model. The mRNA expression of Wnt ligands (Wnt1, Wnt3a, Wnt5a), upstream Wnt signaling regulator genes (Frizzled Receptors 1, 2 and secreted Frizzled related protein sFRP-1) and canonical Wnt intracellular effectors (GSK3ß, Axin1) were assessed by real-time polymerase chain reaction and protein levels of GSK3ß, phospho-GSK3ß (ser 9) by western blotting and localization by immunohistochemistry. The role of GSK3ß in PASMCs proliferation was assessed by overexpression of wild-type GSK3ß (WT) and constitutively active GSK3ß S9A by [3H]-thymidine incorporation assay. Results Increased levels of total and phosphorylated GSK3ß (inhibitory phosphorylation) were observed in lungs and PASMCs isolated from MCT-induced PAH rats compared to controls. Further, stimulation of MCT-PASMCs with growth factors induced GSK3ß inactivation. Most importantly, treatment with the PDGFR inhibitor, Imatinib, attenuated PDGF-BB and FCS induced GSK3ß phosphorylation. Increased expression of GSK3ß observed in lungs and PASMC isolated from MCT-induced PAH rats was confirmed to be clinically relevant as the same observation was identified in human iPAH lung explants. Overexpression of GSK3ß significantly increased MCT-PASMCs proliferation by regulating ERK phosphorylation. Constitutive activation of GSK3ß (GSK3ß S9A, 9th serine replaced to alanine) inhibited MCT-PASMCs proliferation by decreasing ERK phosphorylation. Conclusion This study supports a central role for GSK3ß in vascular remodeling processes and suggests a novel therapeutic opportunity for the treatment of PAH.
Collapse
MESH Headings
- Animals
- Arteries/enzymology
- Arteries/pathology
- Base Sequence
- Blotting, Western
- Cell Proliferation
- Cells, Cultured
- Cloning, Molecular
- DNA Primers
- Disease Models, Animal
- Glycogen Synthase Kinase 3/genetics
- Glycogen Synthase Kinase 3/metabolism
- Glycogen Synthase Kinase 3 beta
- Humans
- Hypertension, Pulmonary/enzymology
- Hypertension, Pulmonary/pathology
- Immunohistochemistry
- Male
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Mutagenesis
- Phosphorylation
- Polymerase Chain Reaction
- Rats
- Wnt Proteins/metabolism
Collapse
Affiliation(s)
| | - Ralph Theo Schermuly
- Medical Clinic II/V, University Hospital, Giessen, Germany
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Xia Tian
- Medical Clinic II/V, University Hospital, Giessen, Germany
| | | | | | | | | | - Werner Seeger
- Medical Clinic II/V, University Hospital, Giessen, Germany
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Soni Savai Pullamsetti
- Medical Clinic II/V, University Hospital, Giessen, Germany
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
- * E-mail:
| |
Collapse
|
23
|
Targeting non-malignant disorders with tyrosine kinase inhibitors. Nat Rev Drug Discov 2011; 9:956-70. [PMID: 21119733 DOI: 10.1038/nrd3297] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Receptor and non-receptor tyrosine kinases are involved in multiple proliferative signalling pathways. Imatinib, one of the first tyrosine kinase inhibitors (TKIs) to be approved, revolutionized the treatment of chronic myelogenous leukaemia, and other TKIs with different spectra of kinase inhibition are used to treat renal cell carcinoma, non-small-cell lung cancer and colon cancer. Studies also support the potential use of TKIs as anti-proliferative agents in non-malignant disorders such as cardiac hypertrophy, and in benign-proliferative disorders including pulmonary hypertension, lung fibrosis, rheumatoid disorders, atherosclerosis, in-stent restenosis and glomerulonephritis. In this Review, we provide an overview of the most recent developments--both experimental as well as clinical--regarding the therapeutic potential of TKIs in non-malignant disorders.
Collapse
|
24
|
Tian J, Fratz S, Hou Y, Lu Q, Görlach A, Hess J, Schreiber C, Datar SA, Oishi P, Nechtman J, Podolsky R, She JX, Fineman JR, Black SM. Delineating the angiogenic gene expression profile before pulmonary vascular remodeling in a lamb model of congenital heart disease. Physiol Genomics 2011; 43:87-98. [PMID: 20978110 PMCID: PMC3026563 DOI: 10.1152/physiolgenomics.00135.2010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 10/20/2010] [Indexed: 01/05/2023] Open
Abstract
Disordered angiogenesis is implicated in pulmonary vascular remodeling secondary to congenital heart diseases (CHD). However, the underlying genes are not well delineated. We showed previously that an ovine model of CHD with increased pulmonary blood flow (PBF, Shunt) has an "angiogenesis burst" between 1 and 4 wk of age. Thus we hypothesized that the increased PBF elicited a proangiogenic gene expression profile before onset of vessel growth. To test this we utilized microarray analysis to identify genes that could be responsible for the angiogenic response. Total RNA was isolated from lungs of Shunt and control lambs at 3 days of age and hybridized to Affymetrix gene chips for microarray analyses (n = 8/group). Eighty-nine angiogenesis-related genes were found to be upregulated and 26 angiogenesis-related genes downregulated in Shunt compared with control lungs (cutting at 1.2-fold difference, P < 0.05). We then confirmed upregulation of proangiogenic genes FGF2, Angiopoietin2 (Angpt2), and Birc5 at mRNA and protein levels and upregulation of ccl2 at mRNA level in 3-day Shunt lungs. Furthermore, we found that pulmonary arterial endothelial cells (PAEC) isolated from fetal lambs exhibited increased expression of FGF2, Angpt2, Birc5, and ccl2 and enhanced angiogenesis when exposed to elevated shear stress (35 dyn/cm²) compared with cells exposed to more physiological shear stress (20 dyn/cm²). Finally, we demonstrated that blocking FGF2, Angpt2, Birc5, or ccl2 signaling with neutralizing antibodies or small interfering RNA (siRNA) significantly decreased the angiogenic response induced by shear stress. In conclusion, we have identified a "proangiogenic" gene expression profile in a lamb model of CHD with increased PBF that precedes onset of pulmonary vascular remodeling. Our data indicate that FGF2, Angpt2, Birc5, and ccl2 may play important roles in the angiogenic response.
Collapse
Affiliation(s)
- Jing Tian
- Vascular Biology Center, Augusta, Georgia 30912, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Adatia I, Kothari SS, Feinstein JA. Pulmonary Hypertension Associated With Congenital Heart Disease. Chest 2010; 137:52S-61S. [DOI: 10.1378/chest.09-2861] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
26
|
Lecht S, Foerster C, Arien-Zakay H, Marcinkiewicz C, Lazarovici P, Lelkes PI. Cardiac microvascular endothelial cells express and release nerve growth factor but not fibroblast growth factor-2. In Vitro Cell Dev Biol Anim 2010; 46:469-76. [DOI: 10.1007/s11626-009-9267-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2009] [Accepted: 11/23/2009] [Indexed: 02/08/2023]
|
27
|
Kulik T, Mullen M, Adatia I. Pulmonary arterial hypertension associated with congenital heart disease. PROGRESS IN PEDIATRIC CARDIOLOGY 2009. [DOI: 10.1016/j.ppedcard.2009.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
28
|
Niu X, Nouraie M, Campbell A, Rana S, Minniti CP, Sable C, Darbari D, Dham N, Reading NS, Prchal JT, Kato GJ, Gladwin MT, Castro OL, Gordeuk VR. Angiogenic and inflammatory markers of cardiopulmonary changes in children and adolescents with sickle cell disease. PLoS One 2009; 4:e7956. [PMID: 19956689 PMCID: PMC2776981 DOI: 10.1371/journal.pone.0007956] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Accepted: 10/26/2009] [Indexed: 11/19/2022] Open
Abstract
Background Pulmonary hypertension and left ventricular diastolic dysfunction are complications of sickle cell disease. Pulmonary hypertension is associated with hemolysis and hypoxia, but other unidentified factors are likely involved in pathogenesis as well. Design and Methods Plasma concentrations of three angiogenic markers (fibroblast growth factor, platelet derived growth factor–BB [PDGF-BB], vascular endothelial growth factor [VEGF]) and seven inflammatory markers implicated in pulmonary hypertension in other settings were determined by Bio-Plex suspension array in 237 children and adolescents with sickle cell disease at steady state and 43 controls. Tricuspid regurgitation velocity (which reflects systolic pulmonary artery pressure), mitral valve E/Edti ratio (which reflects left ventricular diastolic dysfunction), and a hemolytic component derived from four markers of hemolysis and hemoglobin oxygen saturation were also determined. Results Plasma concentrations of interleukin-8, interleukin-10 and VEGF were elevated in the patients with sickle cell disease compared to controls (P≤0.003). By logistic regression, greater values for PDGF-BB (P = 0.009), interleukin-6 (P = 0.019) and the hemolytic component (P = 0.026) were independently associated with increased odds of elevated tricuspid regurgitation velocity while higher VEGF concentrations were associated with decreased odds (P = 0.005) among the patients with sickle cell disease. These findings, which are consistent with reports that PDGF-BB stimulates and VEGF inhibits vascular smooth muscle cell proliferation, did not apply to E/Etdi. Conclusions Circulating concentrations of angiogenic and pro-Inflammatory markers are altered in sickle cell disease children and adolescents with elevated tricuspid regurgitation velocity, a subgroup that may be at risk for developing worsening pulmonary hypertension. Further studies to understand the molecular changes in these children are indicated.
Collapse
Affiliation(s)
- Xiaomei Niu
- Center for Sickle Cell Disease, Howard University, Washington, D. C., United States of America
| | - Mehdi Nouraie
- Center for Sickle Cell Disease, Howard University, Washington, D. C., United States of America
| | - Andrew Campbell
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Sohail Rana
- Center for Sickle Cell Disease, Howard University, Washington, D. C., United States of America
| | - Caterina P. Minniti
- Pulmonary and Vascular Medicine Branch, National Heart, Lung and Blood Institute, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Craig Sable
- Department of Cardiology, Children's National Medical Center, Washington, D. C., United States of America
| | - Deepika Darbari
- Department of Hematology, Children's National Medical Center, Washington, D. C., United States of America
| | - Niti Dham
- Department of Cardiology, Children's National Medical Center, Washington, D. C., United States of America
| | - N. Scott Reading
- University of Utah, ARUP Institute of Clinical and Experimental Pathology, and Veterans Administration Hospital, Salt Lake City, Utah, United States of America
| | - Josef T. Prchal
- University of Utah, ARUP Institute of Clinical and Experimental Pathology, and Veterans Administration Hospital, Salt Lake City, Utah, United States of America
| | - Gregory J. Kato
- Pulmonary and Vascular Medicine Branch, National Heart, Lung and Blood Institute, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mark T. Gladwin
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center and Hemostasis and Vascular Biology Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Oswaldo L. Castro
- Center for Sickle Cell Disease, Howard University, Washington, D. C., United States of America
| | - Victor R. Gordeuk
- Center for Sickle Cell Disease, Howard University, Washington, D. C., United States of America
- * E-mail:
| |
Collapse
|
29
|
|
30
|
Izikki M, Guignabert C, Fadel E, Humbert M, Tu L, Zadigue P, Dartevelle P, Simonneau G, Adnot S, Maitre B, Raffestin B, Eddahibi S. Endothelial-derived FGF2 contributes to the progression of pulmonary hypertension in humans and rodents. J Clin Invest 2009; 119:512-23. [PMID: 19197140 DOI: 10.1172/jci35070] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Accepted: 12/22/2008] [Indexed: 12/29/2022] Open
Abstract
Pulmonary hypertension (PH) is a progressive, lethal lung disease characterized by pulmonary artery SMC (PA-SMC) hyperplasia leading to right-sided heart failure. Molecular events originating in pulmonary ECs (P-ECs) may contribute to the PA-SMC hyperplasia in PH. Thus, we exposed cultured human PA-SMC to medium conditioned by P-EC from patients with idiopathic PH (IPH) or controls and found that IPH P-EC-conditioned medium increased PA-SMC proliferation more than control P-EC medium. Levels of FGF2 were increased in the medium of IPH P-ECs over controls, while there was no detectable difference in TGF-beta1, PDGF-BB, or EGF levels. No difference in FGF2-induced proliferation or FGF receptor type 1 (FGFR1) mRNA levels was detected between IPH and control PA-SMCs. Knockdown of FGF2 in P-EC using siRNA reduced the PA-SMC growth-stimulating effects of IPH P-EC medium by 60% and control P-EC medium by 10%. In situ hybridization showed FGF2 overproduction predominantly in the remodeled vascular endothelium of lungs from patients with IPH. Repeated intravenous FGF2-siRNA administration abolished lung FGF2 production, both preventing and nearly reversing a rat model of PH. Similarly, pharmacological FGFR1 inhibition with SU5402 reversed established PH in the same model. Thus, endothelial FGF2 is overproduced in IPH and contributes to SMC hyperplasia in IPH, identifying FGF2 as a promising target for new treatments against PH.
Collapse
Affiliation(s)
- Mohamed Izikki
- INSERM U841, Faculté de Médecine, 8 avenue du Général Sarrail, 94010 Créteil, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Klein M, Schermuly RT, Ellinghaus P, Milting H, Riedl B, Nikolova S, Pullamsetti SS, Weissmann N, Dony E, Savai R, Ghofrani HA, Grimminger F, Busch AE, Schäfer S. Combined Tyrosine and Serine/Threonine Kinase Inhibition by Sorafenib Prevents Progression of Experimental Pulmonary Hypertension and Myocardial Remodeling. Circulation 2008; 118:2081-90. [DOI: 10.1161/circulationaha.108.779751] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Martina Klein
- From Cardiology Research, Bayer Schering Pharma, Wuppertal (M.K., B.R., A.E.B., S.S.); Max-Planck Institute for Heart and Lung Research, Bad Nauheim (R.T.S.); University of Giessen Lung Center, Giessen (R.T.S., S.N., S.S.P., N.W., E.D., R.S., H.A.G., F.G.); Target Discovery, Bayer Schering Pharma, Wuppertal (P.E.); and Heart and Diabetes Center NRW, Bad Oeynhausen (H.M.), Germany
| | - Ralph T. Schermuly
- From Cardiology Research, Bayer Schering Pharma, Wuppertal (M.K., B.R., A.E.B., S.S.); Max-Planck Institute for Heart and Lung Research, Bad Nauheim (R.T.S.); University of Giessen Lung Center, Giessen (R.T.S., S.N., S.S.P., N.W., E.D., R.S., H.A.G., F.G.); Target Discovery, Bayer Schering Pharma, Wuppertal (P.E.); and Heart and Diabetes Center NRW, Bad Oeynhausen (H.M.), Germany
| | - Peter Ellinghaus
- From Cardiology Research, Bayer Schering Pharma, Wuppertal (M.K., B.R., A.E.B., S.S.); Max-Planck Institute for Heart and Lung Research, Bad Nauheim (R.T.S.); University of Giessen Lung Center, Giessen (R.T.S., S.N., S.S.P., N.W., E.D., R.S., H.A.G., F.G.); Target Discovery, Bayer Schering Pharma, Wuppertal (P.E.); and Heart and Diabetes Center NRW, Bad Oeynhausen (H.M.), Germany
| | - Hendrik Milting
- From Cardiology Research, Bayer Schering Pharma, Wuppertal (M.K., B.R., A.E.B., S.S.); Max-Planck Institute for Heart and Lung Research, Bad Nauheim (R.T.S.); University of Giessen Lung Center, Giessen (R.T.S., S.N., S.S.P., N.W., E.D., R.S., H.A.G., F.G.); Target Discovery, Bayer Schering Pharma, Wuppertal (P.E.); and Heart and Diabetes Center NRW, Bad Oeynhausen (H.M.), Germany
| | - Bernd Riedl
- From Cardiology Research, Bayer Schering Pharma, Wuppertal (M.K., B.R., A.E.B., S.S.); Max-Planck Institute for Heart and Lung Research, Bad Nauheim (R.T.S.); University of Giessen Lung Center, Giessen (R.T.S., S.N., S.S.P., N.W., E.D., R.S., H.A.G., F.G.); Target Discovery, Bayer Schering Pharma, Wuppertal (P.E.); and Heart and Diabetes Center NRW, Bad Oeynhausen (H.M.), Germany
| | - Sevdalina Nikolova
- From Cardiology Research, Bayer Schering Pharma, Wuppertal (M.K., B.R., A.E.B., S.S.); Max-Planck Institute for Heart and Lung Research, Bad Nauheim (R.T.S.); University of Giessen Lung Center, Giessen (R.T.S., S.N., S.S.P., N.W., E.D., R.S., H.A.G., F.G.); Target Discovery, Bayer Schering Pharma, Wuppertal (P.E.); and Heart and Diabetes Center NRW, Bad Oeynhausen (H.M.), Germany
| | - Soni S. Pullamsetti
- From Cardiology Research, Bayer Schering Pharma, Wuppertal (M.K., B.R., A.E.B., S.S.); Max-Planck Institute for Heart and Lung Research, Bad Nauheim (R.T.S.); University of Giessen Lung Center, Giessen (R.T.S., S.N., S.S.P., N.W., E.D., R.S., H.A.G., F.G.); Target Discovery, Bayer Schering Pharma, Wuppertal (P.E.); and Heart and Diabetes Center NRW, Bad Oeynhausen (H.M.), Germany
| | - Norbert Weissmann
- From Cardiology Research, Bayer Schering Pharma, Wuppertal (M.K., B.R., A.E.B., S.S.); Max-Planck Institute for Heart and Lung Research, Bad Nauheim (R.T.S.); University of Giessen Lung Center, Giessen (R.T.S., S.N., S.S.P., N.W., E.D., R.S., H.A.G., F.G.); Target Discovery, Bayer Schering Pharma, Wuppertal (P.E.); and Heart and Diabetes Center NRW, Bad Oeynhausen (H.M.), Germany
| | - Eva Dony
- From Cardiology Research, Bayer Schering Pharma, Wuppertal (M.K., B.R., A.E.B., S.S.); Max-Planck Institute for Heart and Lung Research, Bad Nauheim (R.T.S.); University of Giessen Lung Center, Giessen (R.T.S., S.N., S.S.P., N.W., E.D., R.S., H.A.G., F.G.); Target Discovery, Bayer Schering Pharma, Wuppertal (P.E.); and Heart and Diabetes Center NRW, Bad Oeynhausen (H.M.), Germany
| | - Rajkumar Savai
- From Cardiology Research, Bayer Schering Pharma, Wuppertal (M.K., B.R., A.E.B., S.S.); Max-Planck Institute for Heart and Lung Research, Bad Nauheim (R.T.S.); University of Giessen Lung Center, Giessen (R.T.S., S.N., S.S.P., N.W., E.D., R.S., H.A.G., F.G.); Target Discovery, Bayer Schering Pharma, Wuppertal (P.E.); and Heart and Diabetes Center NRW, Bad Oeynhausen (H.M.), Germany
| | - Hossein A. Ghofrani
- From Cardiology Research, Bayer Schering Pharma, Wuppertal (M.K., B.R., A.E.B., S.S.); Max-Planck Institute for Heart and Lung Research, Bad Nauheim (R.T.S.); University of Giessen Lung Center, Giessen (R.T.S., S.N., S.S.P., N.W., E.D., R.S., H.A.G., F.G.); Target Discovery, Bayer Schering Pharma, Wuppertal (P.E.); and Heart and Diabetes Center NRW, Bad Oeynhausen (H.M.), Germany
| | - Friedrich Grimminger
- From Cardiology Research, Bayer Schering Pharma, Wuppertal (M.K., B.R., A.E.B., S.S.); Max-Planck Institute for Heart and Lung Research, Bad Nauheim (R.T.S.); University of Giessen Lung Center, Giessen (R.T.S., S.N., S.S.P., N.W., E.D., R.S., H.A.G., F.G.); Target Discovery, Bayer Schering Pharma, Wuppertal (P.E.); and Heart and Diabetes Center NRW, Bad Oeynhausen (H.M.), Germany
| | - Andreas E. Busch
- From Cardiology Research, Bayer Schering Pharma, Wuppertal (M.K., B.R., A.E.B., S.S.); Max-Planck Institute for Heart and Lung Research, Bad Nauheim (R.T.S.); University of Giessen Lung Center, Giessen (R.T.S., S.N., S.S.P., N.W., E.D., R.S., H.A.G., F.G.); Target Discovery, Bayer Schering Pharma, Wuppertal (P.E.); and Heart and Diabetes Center NRW, Bad Oeynhausen (H.M.), Germany
| | - Stefan Schäfer
- From Cardiology Research, Bayer Schering Pharma, Wuppertal (M.K., B.R., A.E.B., S.S.); Max-Planck Institute for Heart and Lung Research, Bad Nauheim (R.T.S.); University of Giessen Lung Center, Giessen (R.T.S., S.N., S.S.P., N.W., E.D., R.S., H.A.G., F.G.); Target Discovery, Bayer Schering Pharma, Wuppertal (P.E.); and Heart and Diabetes Center NRW, Bad Oeynhausen (H.M.), Germany
| |
Collapse
|
32
|
Markel TA, Wairiuko GM, Lahm T, Crisostomo PR, Wang M, Herring CM, Meldrum DR. The Right Heart and Its Distinct Mechanisms of Development, Function, and Failure. J Surg Res 2008; 146:304-13. [DOI: 10.1016/j.jss.2007.04.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 03/27/2007] [Accepted: 04/02/2007] [Indexed: 01/21/2023]
|
33
|
Black SM, DeVol JM, Wedgwood S. Regulation of fibroblast growth factor-2 expression in pulmonary arterial smooth muscle cells involves increased reactive oxygen species generation. Am J Physiol Cell Physiol 2007; 294:C345-54. [PMID: 17942638 DOI: 10.1152/ajpcell.00216.2007] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We have previously demonstrated increased fibroblast growth factor-2 (FGF-2) expression in a lamb model of increased pulmonary blood flow secondary to congenital heart disease, which may contribute to the associated increases in pulmonary arterial muscularization. However, the mechanisms underlying these increases in FGF-2 expression remain to be identified. Initially, we found that exogenous FGF-2 increased endogenous FGF-2 promoter activity and protein levels in ovine pulmonary arterial smooth muscle cells (PASMC). Furthermore, we found that these increases in FGF-2 expression were mediated by increases in superoxide levels via NADPH oxidase activation. In addition, FGF-2-mediated increases in FGF-2 expression and PASMC proliferation were attenuated by inhibition of phosphatidylinositol 3-kinase, Akt, and NADPH oxidase. Increases in FGF-2 expression could be stimulated by other factors known to increase reactive oxygen species (ROS) signaling in PASMC (endothelin-1 and transforming growth factor-beta1), whereas antioxidants attenuated these increases. Deletion constructs localized the growth factor- and ROS-sensitive region within the proximal 103 bp of the FGF-2 promoter, and sequence analysis identified a putative hypoxia response element (HRE), a DNA binding site for the ROS-sensitive transcription factor hypoxia-inducible factor-1alpha (HIF-1alpha). Stabilization of HIF-1alpha increased FGF-2 promoter activity, whereas mutation of the putative HRE attenuated FGF-2-induced FGF-2 promoter activity. Furthermore, FGF-2 increased HIF-1alpha protein levels and consensus HRE promoter activity in PASMC via antioxidant-sensitive mechanisms. Thus we conclude that FGF-2 can stimulate its own expression in PASMC via NADPH oxidase-mediated activation of ROS-sensitive transcription factors, including HIF-1alpha. This positive feedback mechanism may contribute to pulmonary vascular remodeling associated with increased pulmonary blood flow.
Collapse
Affiliation(s)
- Stephen M Black
- Vascular Biology Center, Medical College of Georgia, Augusta, Georgia, USA
| | | | | |
Collapse
|