1
|
Yang Z, Li F, Thandavarayan RA, Natarajan K, Martin DR, Li Z, Guha A. Early detection of pulmonary arterial hypertension through [ 18F] positron emission tomography imaging with a vascular endothelial receptor small molecule. Pulm Circ 2024; 14:e12393. [PMID: 39072304 PMCID: PMC11273098 DOI: 10.1002/pul2.12393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 07/30/2024] Open
Abstract
The objective of this study is to provide a positron emission tomography (PET) imaging modality targeting vascular endothelial growth factor receptors (VEGFR) for the early noninvasive detection and assessment of pulmonary arterial hypertension (PAH) severity. To validate the effectiveness of the [18F]VEGFR PET tracer, we utilized a monocrotaline (MCT)-induced PAH rat model. Molecular optical imaging, using a Cy5.5-conjugated VEGFR targeting agent, was employed to demonstrate the uptake of the agent at pulmonary arterioles, correlating with the onset and progression of PAH. Histological examinations of the MCT-PAH rat lung revealed a significant correlation between VEGFR2 expression and the pathogenesis of PAH. Molecular optical imaging demonstrated heightened uptake of the Cy5.5-conjugated VEGFR targeting agent at pulmonary arterioles, corresponding with the onset and progression of PAH. [18F]VEGFR PET showed increased lung uptake detectable in early-stage PAH before increase in pulmonary artery pressures, and this uptake correlated with increased PAH severity. Moreover, when compared to [18F]FDG PET, [18F]VEGFR PET exhibited markedly lower background cardiac signal, enhancing imaging sensitivity for lung abnormalities. Our study provides a compelling evidence for the potential utility of the innovative [18F]VEGFR PET tracer, in non-invasively detecting early signs of PAH, and monitoring its progression. The observed correlations between VEGFR2 expression, molecular optical imaging results, and [18F]VEGFR PET findings support the use of this tracer for early detection, and assessment of PAH severity. The lower background cardiac signal observed with [18F]VEGFR PET further enhances its imaging sensitivity, emphasizing its potential clinical significance.
Collapse
Affiliation(s)
- Zhen Yang
- Department of RadiologyHouston Methodist Academic InstituteHoustonTexasUSA
| | - Feng Li
- Department of RadiologyHouston Methodist Academic InstituteHoustonTexasUSA
| | | | - Kartiga Natarajan
- Department of Cardiovascular SciencesHouston Methodist Research InstituteHoustonTexasUSA
| | - Diego R. Martin
- Department of RadiologyHouston Methodist Academic InstituteHoustonTexasUSA
- Department of RadiologyWeil Cornell MedicineNew YorkNew YorkUSA
| | - Zheng Li
- Department of RadiologyHouston Methodist Academic InstituteHoustonTexasUSA
- Center for Scientific ReviewNIHBethesdaMarylandUSA
| | - Ashrith Guha
- Department of CardiologyHouston Methodist HospitalHoustonTexasUSA
| |
Collapse
|
2
|
Gu S, Goel K, Forbes LM, Kheyfets VO, Yu YRA, Tuder RM, Stenmark KR. Tensions in Taxonomies: Current Understanding and Future Directions in the Pathobiologic Basis and Treatment of Group 1 and Group 3 Pulmonary Hypertension. Compr Physiol 2023; 13:4295-4319. [PMID: 36715285 PMCID: PMC10392122 DOI: 10.1002/cphy.c220010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In the over 100 years since the recognition of pulmonary hypertension (PH), immense progress and significant achievements have been made with regard to understanding the pathophysiology of the disease and its treatment. These advances have been mostly in idiopathic pulmonary arterial hypertension (IPAH), which was classified as Group 1 Pulmonary Hypertension (PH) at the Second World Symposia on PH in 1998. However, the pathobiology of PH due to chronic lung disease, classified as Group 3 PH, remains poorly understood and its treatments thus remain limited. We review the history of the classification of the five groups of PH and aim to provide a state-of-the-art review of the understanding of the pathogenesis of Group 1 PH and Group 3 PH including insights gained from novel high-throughput omics technologies that have revealed heterogeneities within these categories as well as similarities between them. Leveraging the substantial gains made in understanding the genomics, epigenomics, proteomics, and metabolomics of PAH to understand the full spectrum of the complex, heterogeneous disease of PH is needed. Multimodal omics data as well as supervised and unbiased machine learning approaches after careful consideration of the powerful advantages as well as of the limitations and pitfalls of these technologies could lead to earlier diagnosis, more precise risk stratification, better predictions of disease response, new sub-phenotype groupings within types of PH, and identification of shared pathways between PAH and other types of PH that could lead to new treatment targets. © 2023 American Physiological Society. Compr Physiol 13:4295-4319, 2023.
Collapse
Affiliation(s)
- Sue Gu
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
- Cardiovascular Pulmonary Research Lab, University of Colorado School of Medicine, Colorado, USA
- National Jewish Health, Denver, Colorodo, USA
| | - Khushboo Goel
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
- National Jewish Health, Denver, Colorodo, USA
| | - Lindsay M. Forbes
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
| | - Vitaly O. Kheyfets
- Cardiovascular Pulmonary Research Lab, University of Colorado School of Medicine, Colorado, USA
| | - Yen-rei A. Yu
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
- Cardiovascular Pulmonary Research Lab, University of Colorado School of Medicine, Colorado, USA
| | - Rubin M. Tuder
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
- Program in Translational Lung Research, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
| | - Kurt R. Stenmark
- Cardiovascular Pulmonary Research Lab, University of Colorado School of Medicine, Colorado, USA
- Department of Pediatrics Section of Critical Care Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
| |
Collapse
|
3
|
Subedi P, Gasho C, Stembridge M, Williams AM, Patrician A, Ainslie PN, Anholm JD. Pulmonary vascular reactivity to supplemental oxygen in Sherpa and lowlanders during gradual ascent to high altitude. Exp Physiol 2023; 108:111-122. [PMID: 36404588 PMCID: PMC10103769 DOI: 10.1113/ep090458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 10/18/2022] [Indexed: 11/23/2022]
Abstract
NEW FINDINGS What is the central question of this study? How does hypoxic pulmonary vasoconstriction and the response to supplemental oxygen change over time at high altitude? What is the main finding and its importance? Lowlanders and partially de-acclimatized Sherpa both demonstrated pulmonary vascular responsiveness to supplemental oxygen that was maintained for 12 days' exposure to progressively increasing altitude. An additional 2 weeks' acclimatization at 5050 m altitude rendered the pulmonary vasculature minimally responsive to oxygen similar to the fully acclimatized non-ascent Sherpa. Additional hypoxic exposure at that time point did not augment hypoxic pulmonary vasoconstriction. ABSTRACT Prolonged alveolar hypoxia leads to pulmonary vascular remodelling. We examined the time course at altitude, over which hypoxic pulmonary vasoconstriction goes from being acutely reversible to potentially irreversible. Study subjects were lowlanders (n = 20) and two Sherpa groups. All Sherpa were born and raised at altitude. One group (ascent Sherpa, n = 11) left altitude and after de-acclimatization in Kathmandu for ∼7 days re-ascended with the lowlanders over 8-10 days to 5050 m. The second Sherpa group (non-ascent Sherpa, n = 12) remained continuously at altitude. Pulmonary artery systolic pressure (PASP) and pulmonary vascular resistance (PVR) were measured while breathing ambient air and following supplemental oxygen. During ascent PASP and PVR increased in lowlanders and ascent Sherpa; however, with supplemental oxygen, lowlanders had significantly greater decrease in PASP (P = 0.02) and PVR (P = 0.02). After ∼14 days at 5050 m, PASP decreased with supplemental oxygen (mean decrease: 3.9 mmHg, 95% CI 2.1-5.7 mmHg, P < 0.001); however, PVR was unchanged (P = 0.49). In conclusion, PASP and PVR increased with gradual ascent to altitude and decreased via oxygen supplementation in both lowlanders and ascent Sherpa. Following ∼14 days at 5050 m altitude, there was no change in PVR to hypoxia or O2 supplementation in lowlanders or either Sherpa group. These data show that both duration of exposure and residential altitude influence the pulmonary vascular responses to hypoxia.
Collapse
Affiliation(s)
- Prajan Subedi
- Division of PulmonaryCritical Care, Sleep, Hyperbaric Medicine and AllergyDept. of MedicineLoma Linda University School of MedicinePulmonary SectionVA Loma Linda Healthcare SystemLoma LindaCaliforniaUSA
| | - Christopher Gasho
- Division of PulmonaryCritical Care, Sleep, Hyperbaric Medicine and AllergyDept. of MedicineLoma Linda University School of MedicinePulmonary SectionVA Loma Linda Healthcare SystemLoma LindaCaliforniaUSA
| | - Michael Stembridge
- Cardiff School of Sport and Health SciencesCardiff Metropolitan UniversityCardiffUK
| | - Alexandra M. Williams
- Department of Cellular and Physiological SciencesFaculty of MedicineUniversity of British ColumbiaVancouverBCCanada
| | - Alexander Patrician
- Centre for Heart, Lung and Vascular HealthFaculty of Health and Social DevelopmentUniversity of British Columbia – OkanaganKelownaBCCanada
| | - Philip N. Ainslie
- Centre for Heart, Lung and Vascular HealthFaculty of Health and Social DevelopmentUniversity of British Columbia – OkanaganKelownaBCCanada
| | - James D. Anholm
- Division of PulmonaryCritical Care, Sleep, Hyperbaric Medicine and AllergyDept. of MedicineLoma Linda University School of MedicinePulmonary SectionVA Loma Linda Healthcare SystemLoma LindaCaliforniaUSA
| |
Collapse
|
4
|
Malkmus K, Brosien M, Knoepp F, Schaffelhofer L, Grimminger F, Rummel C, Gudermann T, Dietrich A, Birnbaumer L, Weissmann N, Kraut S. Deletion of classical transient receptor potential 1, 3 and 6 alters pulmonary vasoconstriction in chronic hypoxia-induced pulmonary hypertension in mice. Front Physiol 2022; 13:1080875. [PMID: 36569761 PMCID: PMC9768328 DOI: 10.3389/fphys.2022.1080875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic hypoxia-induced pulmonary hypertension (CHPH) is a severe disease that is characterized by increased proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs) leading to pulmonary vascular remodeling. The resulting increase in pulmonary vascular resistance (PVR) causes right ventricular hypertrophy and ultimately right heart failure. In addition, increased PVR can also be a consequence of hypoxic pulmonary vasoconstriction (HPV) under generalized hypoxia. Increased proliferation and migration of PASMCs are often associated with high intracellular Ca2+ concentration. Recent publications suggest that Ca2+-permeable nonselective classical transient receptor potential (TRPC) proteins-especially TRPC1 and 6-are crucially involved in acute and sustained hypoxic responses and the pathogenesis of CHPH. The aim of our study was to investigate whether the simultaneous deletion of TRPC proteins 1, 3 and 6 protects against CHPH-development and affects HPV in mice. We used a mouse model of chronic hypoxia as well as isolated, ventilated and perfused mouse lungs and PASMC cell cultures. Although right ventricular systolic pressure as well as echocardiographically assessed PVR and right ventricular wall thickness (RVWT) were lower in TRPC1, 3, 6-deficient mice, these changes were not related to a decreased degree of pulmonary vascular muscularization and a reduced proliferation of PASMCs. However, both acute and sustained HPV were almost absent in the TRPC1, 3, 6-deficient mice and their vasoconstrictor response upon KCl application was reduced. This was further validated by myographical experiments. Our data revealed that 1) TRPC1, 3, 6-deficient mice are partially protected against development of CHPH, 2) these changes may be caused by diminished HPV and not an altered pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Kathrin Malkmus
- Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Monika Brosien
- Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Fenja Knoepp
- Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Lisa Schaffelhofer
- Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Friedrich Grimminger
- Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Christoph Rummel
- Institute of Veterinary Physiology and Biochemistry, Justus-Liebig-University, Giessen, Germany
| | - Thomas Gudermann
- Walther Straub Institute for Pharmacology and Toxicology, Member of the DZL, Ludwig Maximilians University, Munich, Germany
| | - Alexander Dietrich
- Walther Straub Institute for Pharmacology and Toxicology, Member of the DZL, Ludwig Maximilians University, Munich, Germany
| | - Lutz Birnbaumer
- Institute of Biomedical Research (BIOMED), Catholic University of Argentina, Buenos Aires, Argentina,Laboratory of Signal Transduction, National Institute of Environmental Health Sciences (NIEHS), Durham, United States
| | - Norbert Weissmann
- Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Simone Kraut
- Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany,*Correspondence: Simone Kraut,
| |
Collapse
|
5
|
Shahin Y, Alabed S, Alkhanfar D, Tschirren J, Rothman AMK, Condliffe R, Wild JM, Kiely DG, Swift AJ. Quantitative CT Evaluation of Small Pulmonary Vessels Has Functional and Prognostic Value in Pulmonary Hypertension. Radiology 2022; 305:431-440. [PMID: 35819325 PMCID: PMC9619204 DOI: 10.1148/radiol.210482] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/21/2022] [Accepted: 05/05/2022] [Indexed: 11/11/2022]
Abstract
Background The in vivo relationship between peel pulmonary vessels, small pulmonary vessels, and pulmonary hypertension (PH) is not fully understood. Purpose To quantitatively assess peel pulmonary vessel volumes (PPVVs) and small pulmonary vessel volumes (SPVVs) as estimated from CT pulmonary angiography (CTPA) in different subtypes of PH compared with controls, their relationship to pulmonary function and right heart catheter metrics, and their prognostic value. Materials and Methods In this retrospective single-center study performed from January 2008 to February 2018, quantitative CTPA analysis of total SPVV (TSPVV) (0.4- to 2-mm vessel diameter) and PPVV (within 15, 30, and 45 mm from the lung surface) was performed. Results A total of 1823 patients (mean age, 69 years ± 13 [SD]; 1192 women [65%]) were retrospectively analyzed; 1593 patients with PH (mean pulmonary arterial pressure [mPAP], 43 mmHg ± 13 [SD]) were compared with 230 patient controls (mPAP, 19 mm Hg ± 3). The mean vessel volumes in pulmonary peels at 15-, 30-, and 45-mm depths were higher in pulmonary arterial hypertension (PAH) and PH secondary to lung disease compared with chronic thromboembolic PH (45-mm peel, mean difference: 6.4 mL [95% CI: 1, 11] [P < .001] vs 6.8 mL [95% CI: 1, 12] [P = .01]). Mean small vessel volumes at a diameter of less than 2 mm were lower in PAH and PH associated with left heart disease compared with controls (1.6-mm vessels, mean difference: -4.3 mL [95% CI: -8, -0.1] [P = .03] vs -6.8 mL [95% CI: -11, -2] [P < .001]). In patients with PH, the most significant positive correlation was noted with forced vital capacity percentage predicted (r = 0.30-0.40 [all P < .001] for TSPVVs and r = 0.21-0.25 [all P < .001] for PPVVs). Conclusion The volume of pulmonary small vessels is reduced in pulmonary arterial hypertension and pulmonary hypertension (PH) associated with left heart disease, with similar volume of peel vessels compared with controls. For chronic thromboembolic PH, the volume of peel vessels is reduced. In PH, small pulmonary vessel volume is associated with pulmonary function tests. Clinical trial registration no. NCT02565030 Published under a CC BY 4.0 license Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Yousef Shahin
- From the Department of Infection, Immunity and Cardiovascular Disease
(Y.S., S.A., D.A., A.M.K.R., J.M.W., D.G.K., A.J.S.) and INSIGNEO, Institute for
in silico Medicine (D.G.K., A.J.S.), University of Sheffield, Glossop Rd,
Sheffield S10 2JF, England; Department of Clinical Radiology, Sheffield
Teaching Hospitals, Sheffield, England (Y.S., S.A., A.J.S.); VIDA Diagnostics,
Coralville, Iowa (J.T.); and Sheffield Pulmonary Vascular Disease Unit, Royal
Hallamshire Hospital, Sheffield, England (R.C., D.G.K.)
| | - Samer Alabed
- From the Department of Infection, Immunity and Cardiovascular Disease
(Y.S., S.A., D.A., A.M.K.R., J.M.W., D.G.K., A.J.S.) and INSIGNEO, Institute for
in silico Medicine (D.G.K., A.J.S.), University of Sheffield, Glossop Rd,
Sheffield S10 2JF, England; Department of Clinical Radiology, Sheffield
Teaching Hospitals, Sheffield, England (Y.S., S.A., A.J.S.); VIDA Diagnostics,
Coralville, Iowa (J.T.); and Sheffield Pulmonary Vascular Disease Unit, Royal
Hallamshire Hospital, Sheffield, England (R.C., D.G.K.)
| | - Dheyaa Alkhanfar
- From the Department of Infection, Immunity and Cardiovascular Disease
(Y.S., S.A., D.A., A.M.K.R., J.M.W., D.G.K., A.J.S.) and INSIGNEO, Institute for
in silico Medicine (D.G.K., A.J.S.), University of Sheffield, Glossop Rd,
Sheffield S10 2JF, England; Department of Clinical Radiology, Sheffield
Teaching Hospitals, Sheffield, England (Y.S., S.A., A.J.S.); VIDA Diagnostics,
Coralville, Iowa (J.T.); and Sheffield Pulmonary Vascular Disease Unit, Royal
Hallamshire Hospital, Sheffield, England (R.C., D.G.K.)
| | - Juerg Tschirren
- From the Department of Infection, Immunity and Cardiovascular Disease
(Y.S., S.A., D.A., A.M.K.R., J.M.W., D.G.K., A.J.S.) and INSIGNEO, Institute for
in silico Medicine (D.G.K., A.J.S.), University of Sheffield, Glossop Rd,
Sheffield S10 2JF, England; Department of Clinical Radiology, Sheffield
Teaching Hospitals, Sheffield, England (Y.S., S.A., A.J.S.); VIDA Diagnostics,
Coralville, Iowa (J.T.); and Sheffield Pulmonary Vascular Disease Unit, Royal
Hallamshire Hospital, Sheffield, England (R.C., D.G.K.)
| | - Alex M. K. Rothman
- From the Department of Infection, Immunity and Cardiovascular Disease
(Y.S., S.A., D.A., A.M.K.R., J.M.W., D.G.K., A.J.S.) and INSIGNEO, Institute for
in silico Medicine (D.G.K., A.J.S.), University of Sheffield, Glossop Rd,
Sheffield S10 2JF, England; Department of Clinical Radiology, Sheffield
Teaching Hospitals, Sheffield, England (Y.S., S.A., A.J.S.); VIDA Diagnostics,
Coralville, Iowa (J.T.); and Sheffield Pulmonary Vascular Disease Unit, Royal
Hallamshire Hospital, Sheffield, England (R.C., D.G.K.)
| | - Robin Condliffe
- From the Department of Infection, Immunity and Cardiovascular Disease
(Y.S., S.A., D.A., A.M.K.R., J.M.W., D.G.K., A.J.S.) and INSIGNEO, Institute for
in silico Medicine (D.G.K., A.J.S.), University of Sheffield, Glossop Rd,
Sheffield S10 2JF, England; Department of Clinical Radiology, Sheffield
Teaching Hospitals, Sheffield, England (Y.S., S.A., A.J.S.); VIDA Diagnostics,
Coralville, Iowa (J.T.); and Sheffield Pulmonary Vascular Disease Unit, Royal
Hallamshire Hospital, Sheffield, England (R.C., D.G.K.)
| | - James M. Wild
- From the Department of Infection, Immunity and Cardiovascular Disease
(Y.S., S.A., D.A., A.M.K.R., J.M.W., D.G.K., A.J.S.) and INSIGNEO, Institute for
in silico Medicine (D.G.K., A.J.S.), University of Sheffield, Glossop Rd,
Sheffield S10 2JF, England; Department of Clinical Radiology, Sheffield
Teaching Hospitals, Sheffield, England (Y.S., S.A., A.J.S.); VIDA Diagnostics,
Coralville, Iowa (J.T.); and Sheffield Pulmonary Vascular Disease Unit, Royal
Hallamshire Hospital, Sheffield, England (R.C., D.G.K.)
| | - David G. Kiely
- From the Department of Infection, Immunity and Cardiovascular Disease
(Y.S., S.A., D.A., A.M.K.R., J.M.W., D.G.K., A.J.S.) and INSIGNEO, Institute for
in silico Medicine (D.G.K., A.J.S.), University of Sheffield, Glossop Rd,
Sheffield S10 2JF, England; Department of Clinical Radiology, Sheffield
Teaching Hospitals, Sheffield, England (Y.S., S.A., A.J.S.); VIDA Diagnostics,
Coralville, Iowa (J.T.); and Sheffield Pulmonary Vascular Disease Unit, Royal
Hallamshire Hospital, Sheffield, England (R.C., D.G.K.)
| | - Andrew J. Swift
- From the Department of Infection, Immunity and Cardiovascular Disease
(Y.S., S.A., D.A., A.M.K.R., J.M.W., D.G.K., A.J.S.) and INSIGNEO, Institute for
in silico Medicine (D.G.K., A.J.S.), University of Sheffield, Glossop Rd,
Sheffield S10 2JF, England; Department of Clinical Radiology, Sheffield
Teaching Hospitals, Sheffield, England (Y.S., S.A., A.J.S.); VIDA Diagnostics,
Coralville, Iowa (J.T.); and Sheffield Pulmonary Vascular Disease Unit, Royal
Hallamshire Hospital, Sheffield, England (R.C., D.G.K.)
| |
Collapse
|
6
|
Penumatsa KC, Singhal AA, Warburton RR, Bear MD, Bhedi CD, Nasirova S, Wilson JL, Qi G, Preston IR, Hill NS, Fanburg BL, Kim YB, Toksoz D. Vascular smooth muscle ROCK1 contributes to hypoxia-induced pulmonary hypertension development in mice. Biochem Biophys Res Commun 2022; 604:137-143. [PMID: 35303680 PMCID: PMC9047112 DOI: 10.1016/j.bbrc.2022.02.064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/18/2022] [Indexed: 11/26/2022]
Abstract
Rho kinase (ROCK) is implicated in the development of pulmonary arterial hypertension (PAH) in which abnormal pulmonary vascular smooth muscle (VSM) contractility and remodeling lead to right heart failure. Pharmacologic ROCK inhibitors block experimental pulmonary hypertension (PH) development in rodents but can have off-target effects and do not distinguish between the two ROCK forms, ROCK1 and ROCK2, encoded by separate genes. An earlier study using gene knock out (KO) in mice indicated that VSM ROCK2 is required for experimental PH development, but the role of ROCK1 is not well understood. Here we investigated the in vivo role of ROCK1 in PH development by generating a VSM-targeted homozygous ROCK1 gene KO mouse strain. Adult control mice exposed to Sugen5416 (Su)/hypoxia treatment to induce PH had significantly increased right ventricular systolic pressures (RVSP) and RV hypertrophy versus normoxic controls. In contrast, Su/hypoxia-exposed VSM ROCK1 KO mice did not exhibit significant RVSP elevation, and RV hypertrophy was blunted. Su/hypoxia-induced pulmonary small vessel muscularization was similarly elevated in both control and VSM ROCK1 KO animals. siRNA-mediated ROCK1 knock-down (KD) in human PAH pulmonary arterial SM cells (PASMC) did not affect cell growth. However, ROCK1 KD led to reduced AKT and MYPT1 signaling in serotonin-treated PAH PASMC. The findings suggest that like VSM ROCK2, VSM ROCK1 actively contributes to PH development, but in distinction acts via nonproliferative pathways to promote hypoxemia, and thus may be a distinct therapeutic target in PH.
Collapse
Affiliation(s)
- Krishna C Penumatsa
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Adit A Singhal
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Rod R Warburton
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Michael D Bear
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Chinmayee D Bhedi
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Sabina Nasirova
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Jamie L Wilson
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Guanming Qi
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Ioana R Preston
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Nicholas S Hill
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Barry L Fanburg
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Ave., Boston, MA, 02115, USA
| | - Deniz Toksoz
- Division of Pulmonary, Critical Care and Sleep, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA.
| |
Collapse
|
7
|
Diaz GF, Marquez A, Ruiz-Parra A, Beghetti M, Ivy D. An Acute Hyperoxia Test Predicts Survival in Children with Pulmonary Hypertension Living at High Altitude. High Alt Med Biol 2021; 22:395-405. [PMID: 34905397 PMCID: PMC8742266 DOI: 10.1089/ham.2021.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Diaz, Gabriel F., Alicia Marquez, Ariel Ruiz-Parra, Maurice Beghetti, and Dunbar Ivy. An acute hyperoxia test predicts survival in children with pulmonary hypertension living at high altitude. High Alt Med Biol. 22:395-405, 2021. Background: Pulmonary hypertension (PH) causes significant morbidity and mortality in children at altitude. Materials and Methods: Fifty-two children living at 2,640 m were included. During hyperoxia test (O2Test), patients received high oxygen concentrations (FiO2 >80, through Mask, using Venturi or nonrebreathing mask); echocardiography was used to evaluate pulmonary vasculature reactivity. A decrease >20% from the basal pulmonary artery systolic pressure was considered a positive response. Results: Most of the patients had severe PH. The median age at diagnosis was 4.5 years; 34 were female (65.4%). Idiopathic PH was present in 44 patients (84.6%). Six developed severe PH after ductus closure. They were classified in responders (n = 25), and nonresponders (n = 26). Responders were younger (3 years vs. 7 years, p = 0.02), and 22 (88%), had better functional class (FC) 1-2, than nonresponders: 18 (69.23%) of them had worse FC: 3-4 (p = 0.000). In responders, 10/12 who went to live at low altitude became asymptomatic, compared with 7/13 who remained at high altitude. FC 1-2 was achieved by 70% of the patients with idiopathic PH who went to a low altitude, compared with 30% who continued at high altitude (p = 0.03). In nonresponders, 10/26 patients moved to a low altitude: four improved, one worsened, and five died; of the 16/26 patients living at high altitude, four are stable, eight worsened, and four died. Four patients (30.76%) in responder group and nine (69.24%) in the nonresponder group died (p = 0.03). There were differences between both groups in systolic (88 mm Hg vs. 110 mm Hg; p = 0.037), diastolic (37 mm Hg vs. 56 mm Hg; p = 0.035), and mean pulmonary artery pressures (57 mm Hg vs. 88 mm Hg; p = 0.038). Conclusions: This specific hyperoxia test applied until 24 hours (not published before) helps to predict survival and prognosis of children with PH. Children with PH at a high altitude improve at low altitude.
Collapse
Affiliation(s)
- Gabriel F Diaz
- Department of Pediatrics, Universidad Nacional de Colombia, Fundación Santa Fe de Bogotá, Bogotá Colombia
| | - Alicia Marquez
- Clínica De La Mujer, Centro Policlínico del Olaya, Bogotá, Colombia
| | - Ariel Ruiz-Parra
- Instituto de Investigaciones Clínicas and Department of Obstetrics and Gynecology, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Maurice Beghetti
- Head of Pediatric Cardiology Unit (HUG), Director Pulmonary Hypertension Program (HUG) Children's University Hospital, Geneva, Switzerland
| | - Dunbar Ivy
- Chief and Selby's Chair of Pediatric Cardiology, University of Colorado, School of Medicine, Children's Hospital Colorado, Denver, Colorado, USA
| |
Collapse
|
8
|
Shimoda LA. Cellular Pathways Promoting Pulmonary Vascular Remodeling by Hypoxia. Physiology (Bethesda) 2021; 35:222-233. [PMID: 32490752 DOI: 10.1152/physiol.00039.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Exposure to hypoxia increases pulmonary vascular resistance, leading to elevated pulmonary arterial pressure and, potentially, right heart failure. Vascular remodeling is an important contributor to the increased pulmonary vascular resistance. Hyperproliferation of smooth muscle, endothelial cells, and fibroblasts, and deposition of extracellular matrix lead to increased wall thickness, extension of muscle into normally non-muscular arterioles, and vascular stiffening. This review highlights intrinsic and extrinsic modulators contributing to the remodeling process.
Collapse
Affiliation(s)
- Larissa A Shimoda
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
9
|
Zhao Q, Song P, Zou MH. AMPK and Pulmonary Hypertension: Crossroads Between Vasoconstriction and Vascular Remodeling. Front Cell Dev Biol 2021; 9:691585. [PMID: 34169079 PMCID: PMC8217619 DOI: 10.3389/fcell.2021.691585] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/18/2021] [Indexed: 12/25/2022] Open
Abstract
Pulmonary hypertension (PH) is a debilitating and life-threatening disease characterized by increased blood pressure within the pulmonary arteries. Adenosine monophosphate-activated protein kinase (AMPK) is a heterotrimeric serine-threonine kinase that contributes to the regulation of metabolic and redox signaling pathways. It has key roles in the regulation of cell survival and proliferation. The role of AMPK in PH is controversial because both inhibition and activation of AMPK are preventive against PH development. Some clinical studies found that metformin, the first-line antidiabetic drug and the canonical AMPK activator, has therapeutic efficacy during treatment of early-stage PH. Other study findings suggest the use of metformin is preferentially beneficial for treatment of PH associated with heart failure with preserved ejection fraction (PH-HFpEF). In this review, we discuss the "AMPK paradox" and highlight the differential effects of AMPK on pulmonary vasoconstriction and pulmonary vascular remodeling. We also review the effects of AMPK activators and inhibitors on rescue of preexisting PH in animals and include a discussion of gender differences in the response to metformin in PH.
Collapse
Affiliation(s)
| | | | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
10
|
Liu H, Wang N, Li J, Wang W, Han W, Li Q. AAV1-Mediated shRNA Knockdown of SASH1 in Rat Bronchus Attenuates Hypoxia-Induced Pulmonary Artery Remodeling. Hum Gene Ther 2021; 32:796-805. [PMID: 33297837 DOI: 10.1089/hum.2020.242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Pulmonary hypertension (PH) is a proliferative disease characterized by pulmonary arterial remodeling (PAR). SAM and SH3 domain containing 1 (SASH1) is a novel tumor suppressor gene whose biological function in PH is unclear. In this study, a hypoxia-induced pulmonary hypertension (HPH) rat model was constructed to explore the role of SASH1 in PAR. Histopathological changes in the lung tissue and hemodynamic alteration were detected in SASH1-knockdown rats through adeno-associated virus type-1 (AAV1) infection. In vitro, primary human pulmonary arterial smooth muscle cells (HPASMCs) were transfected with SASH1siRNA to investigate the effects of SASH1 on hypoxia-induced proliferation and migration. The molecular mechanisms associated with SASH1 were explored through knockdown and overexpression approaches. We found that SASH1 expression was significantly increased in rat pulmonary arteries and HPASMCs after hypoxia exposure. In vivo, silencing the SASH1 gene expression improved HPH in rats. The SASH1 downregulation inhibited proliferation and migration of hypoxia-induced HPASMCs. The protein expression of phospho-AKT (known as protein kinase B), proliferating cell nuclear antigen, and matrix metalloproteinase 9 (MMP9) in HPASMCs were increased after SASH1 overexpression, whereas these effects were inhibited by SASH1 knockdown. In conclusion, SASH1 downregulation improved hypoxia-induced PAR and PH. SASH1 may be a novel target for PH gene therapy in the era of precision medicine.
Collapse
Affiliation(s)
- Hong Liu
- Departments of Respiratory and Critical Care Medicine and.,Departments of Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao, China
| | - Ning Wang
- Departments of Respiratory and Critical Care Medicine and
| | - Jun Li
- Departments of Respiratory and Critical Care Medicine and
| | - Wenting Wang
- Departments of Oncology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Wei Han
- Departments of Respiratory and Critical Care Medicine and.,Departments of Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao, China
| | - Qinghai Li
- Departments of Respiratory and Critical Care Medicine and.,Departments of Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao, China
| |
Collapse
|
11
|
Hypoxia and its preconditioning on cardiac and vascular remodelling in experimental animals. Respir Physiol Neurobiol 2020; 285:103588. [PMID: 33253893 DOI: 10.1016/j.resp.2020.103588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 09/28/2020] [Accepted: 11/12/2020] [Indexed: 11/21/2022]
Abstract
Since oxygen (O2) is indispensable for mammalian life, every cell in the body is endowed with mechanisms to detect and to respond to changes in the O2 levels in the microenvironment. The heart and the brain are the two most vital, life-supporting organs requiring a continuous supply of O2 to sustain their high metabolic rate. On being challenged with hypoxia, maintenance of O2 supply to these organs even at the cost of others becomes a priority. This review describes the cardiovascular, skeletal muscle vascular, pulmonary vascular and cerebrovascular remodelling in face of chronic mild hypoxia exposure and the underlying mechanisms, with special reference to the role of oxidative stress, hypoxia signalling, autonomic nervous mechanisms. The significance of the normalized wall index (NWI) in assessing the remodelling of the vessels particularly of the intramyocardial coronary artery has been underscored. The review also highlights the basic concepts of hypoxic preconditioning and the subsequent protection of the brain against an acute ischemic insult in preclinical studies hinting towards its possible therapeutic potential in the management of ischemic stroke.
Collapse
|
12
|
Yan S, Resta TC, Jernigan NL. Vasoconstrictor Mechanisms in Chronic Hypoxia-Induced Pulmonary Hypertension: Role of Oxidant Signaling. Antioxidants (Basel) 2020; 9:E999. [PMID: 33076504 PMCID: PMC7602539 DOI: 10.3390/antiox9100999] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/06/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
Elevated resistance of pulmonary circulation after chronic hypoxia exposure leads to pulmonary hypertension. Contributing to this pathological process is enhanced pulmonary vasoconstriction through both calcium-dependent and calcium sensitization mechanisms. Reactive oxygen species (ROS), as a result of increased enzymatic production and/or decreased scavenging, participate in augmentation of pulmonary arterial constriction by potentiating calcium influx as well as activation of myofilament sensitization, therefore mediating the development of pulmonary hypertension. Here, we review the effects of chronic hypoxia on sources of ROS within the pulmonary vasculature including NADPH oxidases, mitochondria, uncoupled endothelial nitric oxide synthase, xanthine oxidase, monoamine oxidases and dysfunctional superoxide dismutases. We also summarize the ROS-induced functional alterations of various Ca2+ and K+ channels involved in regulating Ca2+ influx, and of Rho kinase that is responsible for myofilament Ca2+ sensitivity. A variety of antioxidants have been shown to have beneficial therapeutic effects in animal models of pulmonary hypertension, supporting the role of ROS in the development of pulmonary hypertension. A better understanding of the mechanisms by which ROS enhance vasoconstriction will be useful in evaluating the efficacy of antioxidants for the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
| | | | - Nikki L. Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (S.Y.); (T.C.R.)
| |
Collapse
|
13
|
Garat CV, Majka SM, Sullivan TM, Crossno JT, Reusch JE, Klemm DJ. CREB depletion in smooth muscle cells promotes medial thickening, adventitial fibrosis and elicits pulmonary hypertension. Pulm Circ 2020; 10:2045894019898374. [PMID: 32313640 PMCID: PMC7158261 DOI: 10.1177/2045894019898374] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 12/11/2019] [Indexed: 01/10/2023] Open
Abstract
Levels of the cAMP-responsive transcription factor, CREB, are reduced in medial smooth muscle cells in remodeled pulmonary arteries from hypertensive calves and rats with chronic hypoxia-induced pulmonary hypertension. Here, we show that chronic hypoxia fails to promote CREB depletion in pulmonary artery smooth muscle cells or elicit significant remodeling of the pulmonary arteries in mice, suggesting that sustained CREB expression prevents hypoxia-induced pulmonary artery remodeling. This hypothesis was tested by generating mice, in which CREB was ablated in smooth muscle cells. Loss of CREB in smooth muscle cells stimulated pulmonary artery thickening, right ventricular hypertrophy, profound adventitial collagen deposition, recruitment of myeloid cells to the adventitia, and elevated right ventricular systolic pressure without exposure to chronic hypoxia. Isolated murine CREB-null smooth muscle cells exhibited serum-independent proliferation and hypertrophy in vitro and medium conditioned by CREB-null smooth muscle cells stimulated proliferation and expression of extracellular matrix proteins by adventitial fibroblasts. We conclude that CREB governs the pathologic switch from homeostatic, quiescent smooth muscle cells to proliferative, synthetic cells that drive arterial remodeling contributing to the development or pulmonary hypertension.
Collapse
Affiliation(s)
- Chrystelle V. Garat
- Cardiovascular Pulmonary Research Laboratory, Department of Medicine, University of Colorado, Aurora, CO, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Susan M. Majka
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Denver, CO, USA
| | - Timothy M. Sullivan
- Cardiovascular Pulmonary Research Laboratory, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Joseph T. Crossno
- Cardiovascular Pulmonary Research Laboratory, Department of Medicine, University of Colorado, Aurora, CO, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Jane E.B. Reusch
- Cardiovascular Pulmonary Research Laboratory, Department of Medicine, University of Colorado, Aurora, CO, USA
- Division of Endocrinology, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Dwight J. Klemm
- Cardiovascular Pulmonary Research Laboratory, Department of Medicine, University of Colorado, Aurora, CO, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO, USA
- Geriatric Research, Education and Clinical Center, Veterans Administration, Eastern Colorado Health Care System, Aurora, CO, USA
| |
Collapse
|
14
|
Tuder RM, Stenmark KR. Perspective: pathobiological paradigms in pulmonary hypertension, time for reappraisal. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1131-L1137. [PMID: 32186206 DOI: 10.1152/ajplung.00067.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
For the past 120 years, there has been a progressive evolution of the pathobiological concepts underlying pulmonary hypertension. Conceptual frameworks, build around the paradigms of excessive vasoconstriction (vs. vasodilation) and, more recently, of the cancer-like hypothesis of pulmonary hypertension, have served to consolidate key discoveries; moreover, they have and continue contributing to innovative advances that have been translated into either successful or potential new therapies. However, those frameworks do not fully address the complexity and challenges facing pulmonary hypertension, particularly those involving the marked heterogeneity of disease presentation and the dynamic changes occurring over time in affected tissues and cells. This is particularly relevant in regards to the molecular pathways of pulmonary hypertension; the ever growing understanding of molecular and cellular pathways requires clarification if they drive distinctive pulmonary vascular lesions in a given lung and disease patients with the same group pulmonary hypertension. Novel methodologies and approaches can start dissecting this key challenge in the field as it is critical to address the key angle of heterogeneity of the disease and reappraisal of disease-modifying therapies.
Collapse
Affiliation(s)
- Rubin M Tuder
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, Aurora, Colorado
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, Division of Critical Care Medicine, Departments of Pediatrics and Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
15
|
The effect of activated κ-opioid receptor (κ-OR) on the role of calcium sensing receptor (CaSR) in preventing hypoxic pulmonary hypertension development. Biomed Pharmacother 2020; 125:109931. [PMID: 32066040 DOI: 10.1016/j.biopha.2020.109931] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 12/10/2019] [Accepted: 12/15/2019] [Indexed: 12/22/2022] Open
Abstract
κ-opioid receptor (κ-OR) plays a key role in preventing hypoxic pulmonary hypertension (HPH) development after activated by exogenous agonist U50,488H. Calcium sensing receptor (CaSR) activation induces HPH by promoting vasoconstriction and vascular remodeling. The activated κ-OR is reported to inhibit the expression of CaSR in pulmonary artery smooth muscle cells (PASMCs). Thus, in this study, we aimed to explore the effect of activated κ-OR on the role of CaSR in preventing HPH development. An HPH rat model was constructed using Sprague-Dawley rats. Changes in mean pulmonary arterial pressure (mPAP) and right ventricular pressure (RVP) mediated by κ-OR agonist U50,488H and CaSR inhibitor NPS2143 were observed. The effects of CaSR agonist spermine and inhibitor NPS2143 on pulmonary artery tension were tested. The expression and localization of κ-OR and CaSR were measured in isolated PASMCs. A cell-counting kit-8 assay was performed to evaluate the effect of spermine in PASMC proliferation. Expression of proliferating cell nuclear antigen (PCNA), Erk, and p-Erk was evaluated by western blot analysis. Results showed that κ-OR and CaSR were co-expressed and colocalized in PASMCs under normoxic and hypoxic conditions. Interactions between κ-OR and CaSR were also observed. Spermine improved vasoconstriction in the pulmonary artery in HPH rats, which was abolished by U50,488H. RVP and mPAP were significantly increased in HPH rats under CaSR stimulation, but were significantly reduced when the rats were pretreated with U50,488H and NPS2143 (P < 0.01). Spermine treatment significantly promoted PASMC proliferation, which was significantly inhibited by U50,488H, p38 inhibitor SB203580, JNK inhibitor SP600125, Erk inhibitor SCH772984, and MEK inhibitor U0126, especially Erk inhibitor (P < 0.01). Spermine significantly increased PCNA and P-Erk expression in hypoxic conditions, which was inhibited by U50,488H and NPS2143. κ-OR stimulation prevented HPH development via the CaSR/MAPK signaling pathway.
Collapse
|
16
|
do Amaral-Silva L, Lambertz M, José Zara F, Klein W, Gargaglioni LH, Bícego KC. Parabronchial remodeling in chicks in response to embryonic hypoxia. ACTA ACUST UNITED AC 2019; 222:jeb.197970. [PMID: 31028104 DOI: 10.1242/jeb.197970] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 04/18/2019] [Indexed: 01/31/2023]
Abstract
The embryonic development of parabronchi occurs mainly during the second half of incubation in precocious birds, which makes this phase sensitive to possible morphological modifications induced by O2 supply limitation. Thus, we hypothesized that hypoxia during the embryonic phase of parabronchial development induces morphological changes that remain after hatching. To test this hypothesis, chicken embryos were incubated entirely (21 days) under normoxia or partially under hypoxia (15% O2 during days 12 to 18). Lung structures, including air capillaries, blood capillaries, infundibula, atria, parabronchial lumen, bronchi, blood vessels larger than capillaries and interparabronchial tissue, in 1- and 10-day-old chicks were analyzed using light microscopy-assisted stereology. Tissue barrier and surface area of air capillaries were measured using electron microscopy-assisted stereology, allowing for calculation of the anatomical diffusion factor. Hypoxia increased the relative volumes of air and blood capillaries, structures directly involved in gas exchange, but decreased the relative volumes of atria in both groups of chicks, and the parabronchial lumen in older chicks. Accordingly, the surface area of the air capillaries and the anatomical diffusion factor were increased under hypoxic incubation. Treatment did not alter total lung volume, relative volumes of infundibula, bronchi, blood vessels larger than capillaries, interparabronchial tissue or the tissue barrier of any group. We conclude that hypoxia during the embryonic phase of parabronchial development leads to a morphological remodeling, characterized by increased volume density and respiratory surface area of structures involved in gas exchange at the expense of structures responsible for air conduction in chicks up to 10 days old.
Collapse
Affiliation(s)
- Lara do Amaral-Silva
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinary Sciences, São Paulo State University, Unesp. Jaboticabal, São Paulo 14884-900, Brazil.,National Institute of Science and Technology - Comparative Physiology (INCT- Fisiologia Comparada), UNESP-Jaboticabal, São Paulo 14884-900, Brazil
| | - Markus Lambertz
- Institut für Zoologie, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany.,Sektion Herpetologie, Zoologisches Forschungsmuseum Alexander Koenig, 53113 Bonn, Germany
| | - Fernando José Zara
- Invertebrate Morphology Lab, Department of Applied Biology, IEAMar and CAUNESP College of Agricultural and Veterinary Sciences, São Paulo State University, Unesp. Jaboticabal, São Paulo 14884-900, Brazil
| | - Wilfried Klein
- National Institute of Science and Technology - Comparative Physiology (INCT- Fisiologia Comparada), UNESP-Jaboticabal, São Paulo 14884-900, Brazil.,Department of Biology, School of Philosophy, Sciences and Literature of Ribeirão Preto, University of São Paulo, São Paulo 14040-901, Brazil
| | - Luciane Helena Gargaglioni
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinary Sciences, São Paulo State University, Unesp. Jaboticabal, São Paulo 14884-900, Brazil.,National Institute of Science and Technology - Comparative Physiology (INCT- Fisiologia Comparada), UNESP-Jaboticabal, São Paulo 14884-900, Brazil
| | - Kênia Cardoso Bícego
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinary Sciences, São Paulo State University, Unesp. Jaboticabal, São Paulo 14884-900, Brazil .,National Institute of Science and Technology - Comparative Physiology (INCT- Fisiologia Comparada), UNESP-Jaboticabal, São Paulo 14884-900, Brazil
| |
Collapse
|
17
|
B Ramachandra A, Humphrey JD. Biomechanical characterization of murine pulmonary arteries. J Biomech 2019; 84:18-26. [PMID: 30598195 PMCID: PMC6361676 DOI: 10.1016/j.jbiomech.2018.12.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 11/11/2018] [Accepted: 12/05/2018] [Indexed: 12/20/2022]
Abstract
The biomechanical properties of the major pulmonary arteries play critical roles in normal physiology as well as in diverse pathophysiologies and clinical interventions. Importantly, advances in medical imaging enable simulations of pulmonary hemodynamics, but such models cannot reach their full potential until they are informed with region-specific material properties. In this paper, we present passive and active biaxial biomechanical data for the right and left main pulmonary arteries from wild-type mice. We also evaluate the suitability of a four-fiber family constitutive model as a descriptor of the passive behavior. Despite regional differences in size, the biaxial mechanical properties, including passive stiffness and elastic energy storage, the biaxial wall stresses at in vivo pressures, and the overall contractile capacity in response to smooth muscle cell stimulation under in vivo conditions are remarkably similar between the right and left branches. The proposed methods and results can serve as baseline protocols and measurements for future biaxial experiments on murine models of pulmonary pathologies, and the constitutive model can inform computational models of normal pulmonary growth and remodeling. Our use of consistent experimental protocols and data analyses can also facilitate comparative studies in health and disease across the systemic and pulmonary circulations as well as studies seeking to understand remodeling in surgeries such as the Fontan procedure, which involves different types of vessels.
Collapse
Affiliation(s)
- Abhay B Ramachandra
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States; Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|
18
|
Mazzuca E, Aliverti A, Miserocchi G. Understanding Vasomotion of Lung Microcirculation by In Vivo Imaging. J Imaging 2019; 5:jimaging5020022. [PMID: 34460470 PMCID: PMC8320900 DOI: 10.3390/jimaging5020022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 11/16/2022] Open
Abstract
The balance of lung extravascular water depends upon the control of blood flow in the alveolar distribution vessels that feed downstream two districts placed in parallel, the corner vessels and the alveolar septal network. The occurrence of an edemagenic condition appears critical as an increase in extravascular water endangers the thinness of the air-blood barrier, thus negatively affecting the diffusive capacity of the lung. We exposed anesthetized rabbits to an edemagenic factor (12% hypoxia) for 120 min and followed by in vivo imaging the micro-vascular morphology through a "pleural window" using a stereo microscope at a magnification of 15× (resolution of 7.2 μm). We measured the change in diameter of distribution vessels (50-200 μm) and corner vessels (<50 μm). On average, hypoxia caused a significant decrease in diameter of both smaller distribution vessels (about ~50%) and corner vessels (about ~25%) at 30 min. After 120 min, reperfusion occurred. Regional differences in perivascular interstitial volume were observed and could be correlated with differences in blood flow control. To understand such difference, we modelled imaged alveolar capillary units, obtained by Voronoi method, integrating microvascular pressure parameters with capillary filtration. Results of the analysis suggested that at 120 min, alveolar blood flow was diverted to the corner vessels in larger alveoli, which were found also to undergo a greater filtration indicating greater proneness to develop lung edema.
Collapse
Affiliation(s)
- Enrico Mazzuca
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milan, Italy
| | - Andrea Aliverti
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milan, Italy
| | - Giuseppe Miserocchi
- Department of Experimental Medicine, Università di Milano Bicocca, 20900 Milan, Italy
- Correspondence: ; Tel.: +39-335-248-452
| |
Collapse
|
19
|
Duerr GD, Feißt A, Halbach K, Verfuerth L, Gestrich C, Wenzel D, Zimmer A, Breuer J, Dewald O. CB2-deficiency is associated with a stronger hypertrophy and remodeling of the right ventricle in a murine model of left pulmonary artery occlusion. Life Sci 2018; 215:96-105. [PMID: 30403990 DOI: 10.1016/j.lfs.2018.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/28/2018] [Accepted: 11/02/2018] [Indexed: 01/10/2023]
Abstract
AIMS Pulmonary hypertension (PH) leads to right ventricular (RV) adaptation and remodeling and has deleterious long-term effects on RV function. The endocannabinoid receptor CB2 has been associated with protective effects in adaptation and remodeling of the left ventricle after ischemia. Therefore, we investigated the role of CB2 receptor in RV adaptation after occlusion of the left pulmonary artery (LPA) in a murine model. MAIN METHODS C57/Bl6 (WT)- and CB2 receptor-deficient (Cnr2-/-)-mice underwent paramedian sternotomy and LPA was occluded using a metal clip. Right heart hemodynamic study (Millar®) preceded organ harvesting for immunohistochemistry and mRNA analysis 7 and 21 days (d) post-occlusion. KEY FINDINGS LPA occlusion led to higher RV systolic pressure in Cnr2-/--hearts, while hemodynamics were comparable with WT-hearts after 21d. Cnr2-/--hearts showed higher macrophage infiltration and lower interleukin-10 expression after 7 d, but otherwise a comparable inflammatory mediator expression profile. Cardiomyocyte-hypertrophy was stronger in Cnr2-/--mice, presenting with higher tenascin-C expression than WT-hearts. Planimetry revealed higher collagen area in Cnr2-/--hearts and small areas of cardiomyocyte-loss. Surrounding cardiomyocytes were cleaved caspase-3- and TUNEL positive in Cnr2-/--hearts. This was associated by maladaptation of myosin heavy-chain isoforms and lower reactive oxygen scavenger enzymes induction in Cnr2-/--hearts. We found comparable morphological changes in both lungs between the two genotypes. SIGNIFICANCE LPA occlusion led to increased systolic pressure and adaptation of RV in CB2-deficient mice. CB2 receptor seems to modulate RV adaptation through expression of contractile elements, reactive oxygen scavenger enzymes, and inflammatory response in order to prevent cardiomyocyte apoptosis.
Collapse
MESH Headings
- Animals
- Apoptosis/physiology
- Disease Models, Animal
- Hypertension, Pulmonary/physiopathology
- Hypertrophy, Right Ventricular/genetics
- Hypertrophy, Right Ventricular/physiopathology
- Inflammation/pathology
- Macrophages/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Cardiac/pathology
- Pulmonary Artery/physiopathology
- Reactive Oxygen Species/metabolism
- Receptor, Cannabinoid, CB2/genetics
- Ventricular Function, Right/physiology
Collapse
Affiliation(s)
- Georg Daniel Duerr
- Department of Cardiac Surgery, University Clinical Center Bonn, Germany.
| | - Andreas Feißt
- Department of Cardiac Surgery, University Clinical Center Bonn, Germany
| | - Katharina Halbach
- Department of Cardiac Surgery, University Clinical Center Bonn, Germany
| | - Luise Verfuerth
- Department of Cardiac Surgery, University Clinical Center Bonn, Germany
| | | | - Daniela Wenzel
- Institute of Physiology I, Life&Brain Center, University of Bonn, Germany
| | - Andreas Zimmer
- Institute of Molecular Psychiatry, Life&Brain Center, University of Bonn, Germany
| | - Johannes Breuer
- Department of Pediatric Cardiology, University Clinical Center Bonn, Germany
| | - Oliver Dewald
- Department of Cardiac Surgery, University Clinical Center Bonn, Germany
| |
Collapse
|
20
|
He S, Lin J, Lin L, Xu Y, Feng J. Shikonin‑mediated inhibition of nestin affects hypoxia‑induced proliferation of pulmonary artery smooth muscle cells. Mol Med Rep 2018; 18:3476-3482. [PMID: 30066896 DOI: 10.3892/mmr.2018.9333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 05/21/2018] [Indexed: 11/06/2022] Open
Abstract
The imbalance between the proliferation and apoptosis of pulmonary artery smooth muscle cells (PASMCs) is of importance in pulmonary vascular remodeling. Shikonin, a naphthoquinone compound extracted from the Chinese medicinal herb Lithospermum erythrorhizon, inhibits the proliferation of rat smooth muscle cells (SMCs). The present study was designed to investigate the effects of shikonin on the proliferation of rat PASMCs and the possible mechanisms involved. Rat PASMCs were cultured under the following five treatment conditions: Normal control; hypoxia for 24 h; hypoxia + 1 µM shikonin for 24 h; hypoxia + 2 µM shikonin for 24 h; and hypoxia + 4 µM shikonin for 24 h. The viability of PASMCs was measured using the Cell Counting Kit‑8 assay, the mRNA expression of nestin (NES) in each group was measured by reverse transcription‑polymerase chain reaction and the protein expression of NES was measured by western blotting. The proliferation of hypoxic PASMCs transfected with NES‑specific small interfering (si)RNA decreased compared with the non‑transfected group. These results indicated that hypoxia induced the proliferation of PASMCs through the enhancement of NES expression. The treatment of hypoxic PASMCs with shikonin resulted in a significant downregulation of NES expression and the inhibition of PASMC proliferation.
Collapse
Affiliation(s)
- Susu He
- Department of Respiratory Medicine, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang 317000, P.R. China
| | - Jian Lin
- Department of Respiratory Medicine, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang 317000, P.R. China
| | - Ling Lin
- Department of Respiratory Medicine, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang 317000, P.R. China
| | - Youzu Xu
- Department of Respiratory Medicine, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang 317000, P.R. China
| | - Jiaxi Feng
- Department of Respiratory Medicine, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang 317000, P.R. China
| |
Collapse
|
21
|
Maston LD, Jones DT, Giermakowska W, Resta TC, Ramiro-Diaz J, Howard TA, Jernigan NL, Herbert L, Maurice AA, Gonzalez Bosc LV. Interleukin-6 trans-signaling contributes to chronic hypoxia-induced pulmonary hypertension. Pulm Circ 2018; 8:2045894018780734. [PMID: 29767573 PMCID: PMC6055240 DOI: 10.1177/2045894018780734] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Interleukin-6 (IL-6) is a pleotropic cytokine that signals through the
membrane-bound IL-6 receptor (mIL-6R) to induce anti-inflammatory
(“classic-signaling”) responses. This cytokine also binds to the soluble IL-6R
(sIL-6R) to promote inflammation (“trans-signaling”). mIL-6R expression is
restricted to hepatocytes and immune cells. Activated T cells release sIL-6R
into adjacent tissues to induce trans-signaling. These cellular actions require
the ubiquitously expressed membrane receptor gp130. Reports show that IL-6 is
produced by pulmonary arterial smooth muscle cells (PASMCs) exposed to hypoxia
in culture as well as the medial layer of the pulmonary arteries in mice exposed
to chronic hypoxia (CH), and IL-6 knockout mice are protected from CH-induced
pulmonary hypertension (PH). IL-6 has the potential to contribute to a broad
array of downstream effects, such as cell growth and migration. CH-induced PH is
associated with increased proliferation and migration of PASMCs to previously
non-muscularized vessels of the lung. We tested the hypothesis that IL-6
trans-signaling contributes to CH-induced PH and arterial remodeling. Plasma
levels of sgp130 were significantly decreased in mice exposed to CH (380 mmHg)
for five days compared to normoxic control mice (630 mmHg), while sIL-6R levels
were unchanged. Consistent with our hypothesis, mice that received the IL-6
trans-signaling-specific inhibitor sgp130Fc, a fusion protein of the soluble
extracellular portion of gp130 with the constant portion of the mouse IgG1
antibody, showed attenuation of CH-induced increases in right ventricular
systolic pressure, right ventricular and pulmonary arterial remodeling as
compared to vehicle (saline)-treated control mice. In addition, PASMCs cultured
in the presence of IL-6 and sIL-6R showed enhanced migration but not
proliferation compared to those treated with IL-6 or sIL-6R alone or in the
presence of sgp130Fc. These results indicate that IL-6 trans-signaling
contributes to pulmonary arterial cell migration and CH-induced PH.
Collapse
Affiliation(s)
- Levi D Maston
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - David T Jones
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Wieslawa Giermakowska
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Juan Ramiro-Diaz
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Tamara A Howard
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Lindsay Herbert
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Anna A Maurice
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Laura V Gonzalez Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
22
|
Hypoxia and Local Inflammation in Pulmonary Artery Structure and Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:325-334. [PMID: 29047096 DOI: 10.1007/978-3-319-63245-2_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hypoxia is recognized as a contributor to pulmonary vascular diseases such as pulmonary hypertension. Hypoxia-induced inflammatory changes can enhance structural and functional changes in pulmonary artery (PA) in the context of PH. Accordingly, understanding how hypoxia and inflammation are linked in the context of pulmonary artery structure and function could be relevant towards development of novel therapies for PH. In this regard, factors such as thymic stromal lymphopoietin (TSLP), an inflammatory cytokine, and brain-derived neurotrophic factor (BDNF), a neurotrophin, have been found critical for nonvascular systems such as airway and asthma. While TSLP canonically affects the immune system, in nonvascular systems, noncanonical effects such as altered [Ca2+]i and cell proliferation have been noted: aspects also relevant to the PA, where there is currently little to no data. Similarly, better known in the nervous system, there is increasing evidence that BDNF is locally produced by structural cells of the airway and can contribute to asthma pathophysiology. In this chapter, we summarize the potential relevance of factors such as TSLP and BDNF to the PA and in the context of hypoxia influences towards development of PH. We focus on cell sources and targets such as PA endothelial cells (PAECs) and smooth muscle cells (PASMCs), and the effects of TSLP or BDNF on intracellular Ca2+ responses to vasoconstrictor agonist, cell proliferation, and potential signaling cascades involved.
Collapse
|
23
|
Huertas A, Guignabert C, Barberà JA, Bärtsch P, Bhattacharya J, Bhattacharya S, Bonsignore MR, Dewachter L, Dinh-Xuan AT, Dorfmüller P, Gladwin MT, Humbert M, Kotsimbos T, Vassilakopoulos T, Sanchez O, Savale L, Testa U, Wilkins MR. Pulmonary vascular endothelium: the orchestra conductor in respiratory diseases. Eur Respir J 2018; 51:13993003.00745-2017. [DOI: 10.1183/13993003.00745-2017] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 02/03/2018] [Indexed: 12/15/2022]
Abstract
The European Respiratory Society (ERS) Research Seminar entitled “Pulmonary vascular endothelium: orchestra conductor in respiratory diseases - highlights from basic research to therapy” brought together international experts in dysfunctional pulmonary endothelium, from basic science to translational medicine, to discuss several important aspects in acute and chronic lung diseases. This review will briefly sum up the different topics of discussion from this meeting which was held in Paris, France on October 27–28, 2016. It is important to consider that this paper does not address all aspects of endothelial dysfunction but focuses on specific themes such as: 1) the complex role of the pulmonary endothelium in orchestrating the host response in both health and disease (acute lung injury, chronic obstructive pulmonary disease, high-altitude pulmonary oedema and pulmonary hypertension); and 2) the potential value of dysfunctional pulmonary endothelium as a target for innovative therapies.
Collapse
|
24
|
Mu YP, Huang QH, Zhu JL, Zheng SY, Yan FR, Zhuang XL, Sham JSK, Lin MJ. Magnesium attenuates endothelin-1-induced vasoreactivity and enhances vasodilatation in mouse pulmonary arteries: Modulation by chronic hypoxic pulmonary hypertension. Exp Physiol 2018; 103:604-616. [PMID: 29363240 DOI: 10.1113/ep086655] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/17/2018] [Indexed: 12/28/2022]
Abstract
NEW FINDINGS What is the central question of this study? The central goal of this study was to elucidate the role of magnesium in the regulation of pulmonary vascular reactivity in relationship to hypoxic pulmonary hypertension. What is the main finding and its importance? We found that magnesium is essential for normal vasoreactivity of the pulmonary artery. Increasing the magnesium concentration attenuates vasoconstriction and improves vasodilatation via release of nitric oxide. Pulmonary hypertension is associated with endothelial dysfunction resulting in the suppression of magnesium modulation of vasodilatation. These results provide evidence that magnesium is important for the modulation of pulmonary vascular function. ABSTRACT Pulmonary hypertension (PH) is characterized by enhanced vasoreactivity and sustained pulmonary vasoconstriction, arising from aberrant Ca2+ homeostasis in pulmonary arterial (PA) smooth muscle cells. In addition to Ca2+ , magnesium, the most abundant intracellular divalent cation, also plays crucial roles in many cellular processes that regulate cardiovascular function. Recent findings suggest that magnesium regulates vascular functions by altering the vascular responses to vasodilator and vasoactive agonists and affects endothelial function by modulating endothelium-dependent vasodilatation in hypertension. Administration of magnesium also decreased pulmonary arterial pressure and improved cardiac output in animal models of PH. However, the role of magnesium in the regulation of pulmonary vascular function related to PH has not been studied. In this study, we examined the effects of magnesium on endothelin-1 (ET-1)-induced vasoconstriction, ACh-induced vasodilatation and the generation of NO in PAs of normoxic mice and chronic hypoxia (CH)-treated mice. Our data showed that removal of extracellular magnesium suppressed vasoreactivity of PAs to both ET-1 and ACh. A high concentration of magnesium (4.8 mm) inhibited ET-1-induced vasoconstriction in endothelium-intact or endothelium-disrupted PAs of normoxic and CH-treated mice, and enhanced the ACh-induced production of NO in PAs of normoxic mice. Moreover, magnesium enhanced ACh-induced vasodilatation in PAs of normoxic mice, and the enhancement was completely abolished after exposure to CH. Hence, in this study we demonstrated that increasing the magnesium concentration can attenuate the ET-1-induced contractile response and improve vasodilatation via release of NO from the endothelium. We also demonstrated that chronic exposure to hypoxia can cause endothelial dysfunction resulting in suppression of the magnesium-dependent modulation of vasodilatation.
Collapse
Affiliation(s)
- Yun-Ping Mu
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Fujian Medical University, Fuzhou, Fujian, PR China.,Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Qiu-Hong Huang
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Fujian Medical University, Fuzhou, Fujian, PR China.,Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Jie-Ling Zhu
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Fujian Medical University, Fuzhou, Fujian, PR China.,Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Si-Yi Zheng
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Fujian Medical University, Fuzhou, Fujian, PR China.,Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Fu-Rong Yan
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Fujian Medical University, Fuzhou, Fujian, PR China.,Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Xiao-Ling Zhuang
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Fujian Medical University, Fuzhou, Fujian, PR China.,Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| | - James S K Sham
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Fujian Medical University, Fuzhou, Fujian, PR China.,Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mo-Jun Lin
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Fujian Medical University, Fuzhou, Fujian, PR China.,Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| |
Collapse
|
25
|
Chen J, Wang YX, Dong MQ, Zhang B, Luo Y, Niu W, Li ZC. Reoxygenation Reverses Hypoxic Pulmonary Arterial Remodeling by Inducing Smooth Muscle Cell Apoptosis via Reactive Oxygen Species-Mediated Mitochondrial Dysfunction. J Am Heart Assoc 2017. [PMID: 28645933 PMCID: PMC5669176 DOI: 10.1161/jaha.117.005602] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Pulmonary arterial remodeling, a main characteristic of hypoxic pulmonary hypertension, can gradually reverse once oxygen has been restored. Previous studies documented that apoptosis increased markedly during the reversal of remodeled pulmonary arteries, but the types of cells and mechanisms related to the apoptosis have remained elusive. This study aimed to determine whether pulmonary artery smooth muscle cell (PASMC)‐specific apoptosis was involved in the reoxygenation‐induced reversal of hypoxic pulmonary arterial remodeling and elucidate the underlying mechanism. Methods and Results Hypoxic pulmonary hypertension was induced in adult male Sprague‐Dawley rats (n=6/group) by chronic hypobaric hypoxia. and the hypoxic pulmonary hypertension rats were then transferred to a normoxia condition. During reoxygenation, hypoxia‐induced pulmonary arterial remodeling gradually reversed. The reversal of remodeled pulmonary arteries was associated with increased H2O2 and with changes in lung expression of cleaved caspase3/PARP, Bax, and Bcl‐2, consistent with increased apoptosis. The PASMC apoptosis, in particular, increased remarkably during this reversal. In vitro, reoxygenation induced the apoptosis of cultured rat primary PASMCs accompanied by increased mitochondrial reactive oxygen species, mitochondrial dysfunction, and the release of cytochrome C from mitochondria to cytoplasm. Clearance of reactive oxygen species alleviated mitochondrial dysfunction as well as the release of cytochrome C and, finally, decreased PASMC apoptosis. Conclusions Reoxygenation‐induced apoptosis of PASMCs is implicated in the reversal of hypoxic pulmonary arterial remodeling, which may be attributed to the mitochondrial reactive oxygen species–mediated mitochondrial dysfunction.
Collapse
MESH Headings
- Animals
- Apoptosis
- Apoptosis Regulatory Proteins/metabolism
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/physiopathology
- Hypoxia/complications
- Hypoxia/metabolism
- Hypoxia/pathology
- Hypoxia/physiopathology
- Male
- Mitochondria, Muscle/metabolism
- Mitochondria, Muscle/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Oxygen/metabolism
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Signal Transduction
- Time Factors
- Vascular Remodeling
Collapse
Affiliation(s)
- Jian Chen
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Yan-Xia Wang
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Ming-Qing Dong
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Bo Zhang
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Ying Luo
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Wen Niu
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Zhi-Chao Li
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
26
|
Rodrigues MP, Vissoci CM, Rosa SP, Negreiros SBC. 24-Hour Hypoxia and Pulmonary Hypertension in Patients with Idiopathic Pulmonary Fibrosis. Open Respir Med J 2017; 11:10-16. [PMID: 28659997 PMCID: PMC5470069 DOI: 10.2174/1874306401711010010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/03/2017] [Accepted: 03/03/2017] [Indexed: 01/09/2023] Open
Abstract
Background: The quantification of hypoxia based on resting partial pressure of arterial oxygen (PaO2) may underestimate hypoxia related to activities of daily living or sleep and thus not accurately reflect pulmonary hypertension (PH). The aim of the present study was to investigate the association of resting PaO2 with percent time of SpO2 below 90% (T90) and 88% (T88) in 24 hours. We also evaluated the capacity of hypoxia measures to predict PH in patients with idiopathic pulmonary fibrosis (IPF). Method: This cross-sectional study included 27 patients with IPF presenting PaO2 ≥ 55 mmHg and not receiving home oxygen therapy. All were submitted to blood gas measurement, 24-h oximetry, and transthoracic Doppler echocardiography to estimate systolic pulmonary artery pressure (SPAP). Patients were divided into three groups according to resting PaO2: 55-55.9 mmHg (A); 60-60.9 mmHg (B); ≥ 70 mmHg (C). PH was defined as “likely” if SPAP > 50 mmHg, and as possible for SPAP between 37 and 50 mmHg. Results: T90 and T88 in Groups A, B, and C were as follows: 59.9±29% and 44.1±34%; 49.3±34% and 29.9±31%; 17.1±25% and 8.8±18% respectively, with significant differences between the groups for both T90 (p ≤ 0.01) and T88 (p = 0.02). PaO2 was inversely correlated with T90 (r = -0.398; p = 0.04) and T88 (r = -0.351; p = 0.07). Hypoxia variables did not correlate with SPAP, and were not able to predict PH. Conclusion: Percent time of SpO2 below 90% and 88% in 24 hours revealed periods of severe hypoxia even in patients with borderline-normal resting PaO2. However, none of the present hypoxia variables was capable of predicting PH.
Collapse
Affiliation(s)
- Marcelo P Rodrigues
- Department of Pulmonology, School of Medicine, Universidade de Brasília (UnB), Brasília, DF, Brazil
| | | | - Samuel P Rosa
- Department of Cardiology, Hospital de Base, Brasília, DF, Brazil
| | - Sandra B C Negreiros
- Department of Pulmonology, School of Medicine, Universidade de Brasília (UnB), Brasília, DF, Brazil
| |
Collapse
|
27
|
Wang Y, Ma TT, Gao NN, Zhou XL, Jiang H, Guo R, Jia LN, Chang H, Gao Y, Gao ZM, Pan L. Effect of Tongxinluo on pulmonary hypertension and pulmonary vascular remodeling in rats exposed to a low pressure hypoxic environment. JOURNAL OF ETHNOPHARMACOLOGY 2016; 194:668-673. [PMID: 27737815 DOI: 10.1016/j.jep.2016.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 09/27/2016] [Accepted: 10/03/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tongxinluo (TXL), which is a Chinese medicine rooted from traditional used herbs, has been used in clinic to treat cardiovascular and cerebrovascular diseases. However, it remains unknown whether TXL alleviates low pressure hypoxic pulmonary hypertension. AIM OF THE STUDY Here, we aimed to observe the influence of TXL on pulmonary hypertension in a rat model that exposed to high altitude environment characterized by low pressure hypoxia. MATERIALS AND METHODS A total of 32 male Sprague-Dawley rats were divided into four groups: control group (normal pressure and normoxia), pulmonary hypertension group (PAH, the parameter is equal to that in altitude 5000m), TXL group (rats living in environment equal to that at altitude of 5000m received TXL treatment), vardenafil group (VDNF, rats living in environment equal to that altitude of 5000m received vardenafil treatment). The high altitude environment was created in chamber by adjusting the inner pressure and oxygen content concomitantly. Before entering the chamber, the TXL group was given TXL (1.2gkg-1d-1) for 28 days, and the VDNF group was given VDNF (0.1gkg-1d-1) for 28 days. After 28 days, the mean pulmonary artery pressure (mPAP) and right ventricular pressure was measured using right heart catheterization. The weight of the right ventricle (RV), left ventricle (LV) and interventricular septum (IVS) was measured, and the right ventricular mass index was calculated. Lung tissue was subjected to hematoxylin and elastic fiber staining, and the medial wall thickness (MT), medial wall cross-sectional area (MA), MT%, and MA% were measured. Proliferative activity within the pulmonary arteries was quantified by Ki67staining. RESULTS After 28 days, as compared with that in normal control group, animals living in the chamber (PAH group) showed a significant increase in mPAP( 47.5mmHg versus 18mmHg), RV/LV+IVS (0.45 versus 0.21) and MA% (78% versus 44%), respectively. Administration of TXL resulted in a significant decrease of 20mmHg in mPAP, returning of RV/LV+IVS to 0.27, and a 40% reduction in MT% compared with that in PAH group. In the VDNF group, RV/LV+IVS and MT% was 0.268 and 38.77, significantly lower than that in PAH group. While, mPAP increased by 12.5mmHg with treatment by VDNF. In contrast to the PAH group, alpha- Smooth Muscle Actin (α-SMA reduced by 19% in the TXL group (p=0.005) and 16% in the VDNF group (p=0.01). Ki67 expression in the VDNF group was significantly lower than the PAH group (P<0.01). Ki67 expression in the TXL group was significantly lower than the PAH group (P<0.01). Compared with the VDNF group, the indexes above reduced in the TXL group. Our results indicate that TXL significantly reduces pulmonary artery pressure, right ventricular hypertrophy, pulmonary small artery wall thickness, and luminal stenosis. In addition, smooth muscle proliferation markedly decreased and muscular artery decreased. However, TXL was unable to restore parameters to control levels. CONCLUSIONS The automatic-adjusted low pressure hypoxic chamber was capable of establishing a pulmonary hypertension rat model at an altitude of 5000m. Compared with VDNF, TXL decreased mPAP and right ventricular hypertrophy index (RVHI) in the pulmonary hypertension rat model, and prevented vascular remodeling by reducing small pulmonary artery thickening, smooth muscle thickening and proliferation. Thus, TXL may be a potential treatment for pulmonary hypertension.
Collapse
Affiliation(s)
- Yong Wang
- Department of Geriatric Medicine, Beijing Shijitan Hospital Affiliated to Capital Medical University; Hypoxia Laboratory, Beijing Shijitan Hospital Affiliated to Capital Medical University
| | - Ting-Ting Ma
- Department of Geriatric Medicine, Beijing Shijitan Hospital Affiliated to Capital Medical University; Hypoxia Laboratory, Beijing Shijitan Hospital Affiliated to Capital Medical University
| | - Na-Na Gao
- Central Laboratory, Beijing Shijitan Hospital Affiliated to Capital Medical University
| | - Xiao-Ling Zhou
- Department of Geriatric Medicine, Beijing Shijitan Hospital Affiliated to Capital Medical University; Hypoxia Laboratory, Beijing Shijitan Hospital Affiliated to Capital Medical University
| | - Hong Jiang
- Department of Geriatric Medicine, Beijing Shijitan Hospital Affiliated to Capital Medical University; Hypoxia Laboratory, Beijing Shijitan Hospital Affiliated to Capital Medical University
| | - Rui Guo
- Department of Geriatric Medicine, Beijing Shijitan Hospital Affiliated to Capital Medical University; Hypoxia Laboratory, Beijing Shijitan Hospital Affiliated to Capital Medical University
| | - Li-Na Jia
- Department of Geriatric Medicine, Beijing Shijitan Hospital Affiliated to Capital Medical University; Hypoxia Laboratory, Beijing Shijitan Hospital Affiliated to Capital Medical University
| | - Hong Chang
- Department of the Pathology, Beijing Shijitan Hospital Affiliated to Capital Medical University
| | - Ying Gao
- Department of the Pathology, Beijing Shijitan Hospital Affiliated to Capital Medical University
| | - Zhi-Min Gao
- Central Laboratory, Beijing Shijitan Hospital Affiliated to Capital Medical University
| | - Lei Pan
- Department of Geriatric Medicine, Beijing Shijitan Hospital Affiliated to Capital Medical University; Hypoxia Laboratory, Beijing Shijitan Hospital Affiliated to Capital Medical University.
| |
Collapse
|
28
|
Upregulation of MicroRNA-214 Contributes to the Development of Vascular Remodeling in Hypoxia-induced Pulmonary Hypertension Via Targeting CCNL2. Sci Rep 2016; 6:24661. [PMID: 27381447 PMCID: PMC4933872 DOI: 10.1038/srep24661] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 03/03/2016] [Indexed: 11/08/2022] Open
Abstract
Hypoxia-induced pulmonary hypertension (PH), which is characterized by vascular remodeling of blood vessels, is a significant complication of chronic obstructive pulmonary disease (COPD). In this study, we screened 13 candidate miRNAs in pulmonary artery smooth muscle cells (PASMCs) harvested from COPD patients with PH (n = 18) and normal controls (n = 15) and found that the expression of miR-214 was differentially expressed between these two groups. Additionally, cyclin L2 (CCNL2) was validated as a target of miR-214 in PASMCs using a luciferase assay. Based on real-time PCR, immunohistochemistry and western blot, the expression of CCNL2 was substantially downregulated in PASMCs from COPD patients with PH compared with those from normal controls. Moreover, the relationship between miRNA and mRNA expression was confirmed using real-time PCR and western blot in PASMCs transfected with miR-214 mimics. Furthermore, the introduction of miR-214 significantly promoted the proliferation of PASMCs by suppressing cell apoptosis, and this effect was mediated by the downregulation of CCNL2. Exposure of PASMCs to hypoxia significantly increased the expression of miR-214, decreased the expression of CCNL2, and promoted cell proliferation. However, these effects were significantly attenuated by the introduction of miR-214 inhibitors, which significantly downregulated miR-214 expression and upregulated CCNL2 expression.
Collapse
|
29
|
Abstract
The circulation of the lung is unique both in volume and function. For example, it is the only organ with two circulations: the pulmonary circulation, the main function of which is gas exchange, and the bronchial circulation, a systemic vascular supply that provides oxygenated blood to the walls of the conducting airways, pulmonary arteries and veins. The pulmonary circulation accommodates the entire cardiac output, maintaining high blood flow at low intravascular arterial pressure. As compared with the systemic circulation, pulmonary arteries have thinner walls with much less vascular smooth muscle and a relative lack of basal tone. Factors controlling pulmonary blood flow include vascular structure, gravity, mechanical effects of breathing, and the influence of neural and humoral factors. Pulmonary vascular tone is also altered by hypoxia, which causes pulmonary vasoconstriction. If the hypoxic stimulus persists for a prolonged period, contraction is accompanied by remodeling of the vasculature, resulting in pulmonary hypertension. In addition, genetic and environmental factors can also confer susceptibility to development of pulmonary hypertension. Under normal conditions, the endothelium forms a tight barrier, actively regulating interstitial fluid homeostasis. Infection and inflammation compromise normal barrier homeostasis, resulting in increased permeability and edema formation. This article focuses on reviewing the basics of the lung circulation (pulmonary and bronchial), normal development and transition at birth and vasoregulation. Mechanisms contributing to pathological conditions in the pulmonary circulation, in particular when barrier function is disrupted and during development of pulmonary hypertension, will also be discussed.
Collapse
Affiliation(s)
- Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Larissa A. Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
30
|
Siemsen DW, Dobrinen E, Han S, Chiocchi K, Meissner N, Swain SD. Vascular Dysfunction in Pneumocystis-Associated Pulmonary Hypertension Is Related to Endothelin Response and Adrenomedullin Concentration. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 186:259-69. [PMID: 26687815 DOI: 10.1016/j.ajpath.2015.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/29/2015] [Accepted: 10/22/2015] [Indexed: 10/22/2022]
Abstract
Pulmonary hypertension subsequent to an infectious disease can be due to vascular structural remodeling or to functional alterations within various vascular cell types. In our previous mouse model of Pneumocystis-associated pulmonary hypertension, we found that vascular remodeling was not responsible for observed increases in right ventricular pressures. Here, we report that the vascular dysfunction we observed could be explained by an enhanced response to endothelin-1 (20% greater reduction in lumen diameter, P ≤ 0.05), corresponding to an up-regulation of similar magnitude (P ≤ 0.05) of the endothelin A receptor in the lung tissue. This effect was potentially augmented by a decrease in production of the pulmonary vasodilator adrenomedullin of almost 70% (P ≤ 0.05). These changes did not occur in interferon-γ knockout mice similarly treated, which do not develop pulmonary hypertension under these circumstances. Surprisingly, we did not observe any relevant changes in the vascular endothelial nitric oxide synthase vasodilatory response, which is a common potential site of inflammatory alterations to pulmonary vascular function. Our results indicate the diverse mechanisms by which inflammatory responses to prior infections can cause functionally relevant changes in vascular responses in the lung, promoting the development of pulmonary hypertension.
Collapse
Affiliation(s)
- Dan W Siemsen
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Erin Dobrinen
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Soo Han
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Kari Chiocchi
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Nicole Meissner
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Steve D Swain
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana.
| |
Collapse
|
31
|
Postolow F, Fediuk J, Nolette N, Hinton M, Dakshinamurti S. Thromboxane promotes smooth muscle phenotype commitment but not remodeling of hypoxic neonatal pulmonary artery. FIBROGENESIS & TISSUE REPAIR 2015; 8:20. [PMID: 26583045 PMCID: PMC4650498 DOI: 10.1186/s13069-015-0037-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 10/20/2015] [Indexed: 12/19/2022]
Abstract
Background Persistent pulmonary hypertension of the newborn (PPHN) is characterized by vasoconstriction and pulmonary vascular remodeling. Remodeling is believed to be a response to physical or chemical stimuli including pro-mitotic inflammatory mediators such as thromboxane. Our objective was to examine the effects of hypoxia and thromboxane signaling ex vivo and in vitro on phenotype commitment, cell cycle entry, and proliferation of PPHN and control neonatal pulmonary artery (PA) myocytes in tissue culture. Methods To examine concurrent effects of hypoxia and thromboxane on myocyte growth, serum-fed first-passage newborn porcine PA myocytes were randomized into normoxic (21 % O2) or hypoxic (10 % O2) culture for 3 days, with daily addition of thromboxane mimetic U46619 (10−9 to 10−5 M) or diluent. Cell survival was detected by MTT assay. To determine the effect of chronic thromboxane exposure (versus whole serum) on activation of arterial remodeling, PPHN was induced in newborn piglets by a 3-day hypoxic exposure (FiO2 0.10); controls were 3 day-old normoxic and day 0 piglets. Third-generation PA were segmented and cultured for 3 days in physiologic buffer, Ham’s F-12 media (in the presence or absence of 10 % fetal calf serum), or media with 10−6 M U46619. DNA synthesis was measured by 3H-thymidine uptake, protein synthesis by 3H-leucine uptake, and proliferation by immunostaining for Ki67. Cell cycle entry was studied by laser scanning cytometry of nuclei in arterial tunica media after propidium iodide staining. Phenotype commitment was determined by immunostaining tunica media for myosin heavy chain and desmin, quantified by laser scanning cytometry. Results Contractile and synthetic myocyte subpopulations had differing responses to thromboxane challenge. U46619 decreased proliferation of synthetic and contractile myocytes. PPHN arteries exhibited decreased protein synthesis under all culture conditions. Serum-supplemented PA treated with U46619 had decreased G1/G0 phase myocytes and an increase in S and G2/M. When serum-deprived, PPHN PA incubated with U46619 showed arrested cell cycle entry (increased G0/G1, decreased S and G2/M) and increased abundance of contractile phenotype markers. Conclusions We conclude that thromboxane does not initiate phenotypic dedifferentiation and proliferative activation in PPHN PA. Exposure to thromboxane triggers cell cycle exit and myocyte commitment to contractile phenotype.
Collapse
Affiliation(s)
- Fabiana Postolow
- Department of Pediatrics, University of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada
| | - Jena Fediuk
- Department of Physiology, University of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada ; Biology of Breathing Group, Manitoba Institute of Child Health, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada
| | - Nora Nolette
- Biology of Breathing Group, Manitoba Institute of Child Health, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada
| | - Martha Hinton
- Biology of Breathing Group, Manitoba Institute of Child Health, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada
| | - Shyamala Dakshinamurti
- Department of Pediatrics, University of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada ; Department of Physiology, University of Manitoba, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada ; Biology of Breathing Group, Manitoba Institute of Child Health, 715 McDermot Avenue, Winnipeg, MB R3E 3P4 Canada ; Section of Neonatology, WS012 Women's Hospital, 735 Notre Dame Ave, Winnipeg, MB R3E 0L8 Canada
| |
Collapse
|
32
|
Kandhi S, Qin J, Froogh G, Jiang H, Luo M, Wolin MS, Huang A, Sun D. EET-dependent potentiation of pulmonary arterial pressure: sex-different regulation of soluble epoxide hydrolase. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1478-86. [PMID: 26498250 DOI: 10.1152/ajplung.00208.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 10/17/2015] [Indexed: 01/24/2023] Open
Abstract
We tested the hypothesis that suppression of epoxyeicosatrienoic acid (EET) metabolism via genetic knockout of the gene for soluble epoxide hydrolase (sEH-KO), or female-specific downregulation of sEH expression, plays a role in the potentiation of pulmonary hypertension. We used male (M) and female (F) wild-type (WT) and sEH-KO mice; the latter have high pulmonary EETs. Right ventricular systolic pressure (RVSP) and mean arterial blood pressure (MABP) in control and in response to in vivo administration of U46619 (thromboxane analog), 14,15-EET, and 14,15-EEZE [14,15-epoxyeicosa-5(z)-enoic acid; antagonist of EETs] were recorded. Basal RVSP was comparable among all groups of mice, whereas MABP was significantly lower in F-WT than M-WT mice and further reduced predominantly in F-KO compared with M-KO mice. U46619 dose dependently increased RVSP and MABP in all groups of mice. The increase in RVSP was significantly greater and coincided with smaller increases in MABP in M-KO and F-WT mice compared with M-WT mice. In F-KO mice, the elevation of RVSP by U46619 was even higher than in M-KO and F-WT mice, associated with the least increase in MABP. 14,15-EEZE prevented the augmentation of U46619-induced elevation of RVSP in sEH-KO mice, whereas 14,15-EET-induced pulmonary vasoconstriction was comparable in all groups of mice. sEH expression in the lungs was reduced, paralleled with higher levels of EETs in F-WT compared with M-WT mice. In summary, EETs initiate pulmonary vasoconstriction but act as vasodilators systemically. High pulmonary EETs, as a function of downregulation or deletion of sEH, potentiate U46619-induced increases in RVSP in a female-susceptible manner.
Collapse
Affiliation(s)
- Sharath Kandhi
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Jun Qin
- Department of Physiology, New York Medical College, Valhalla, New York; Department of Surgery, Renji Hospital, Shanghai Jiaotong University, School of Medicine, People's Republic of China; and
| | - Ghezal Froogh
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Houli Jiang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Meng Luo
- Department of Surgery, Renji Hospital, Shanghai Jiaotong University, School of Medicine, People's Republic of China; and Shanghai 9th Hospital, Shanghai Jiaotong University, School of Medicine, People's Republic of China
| | - Michael S Wolin
- Department of Physiology, New York Medical College, Valhalla, New York
| | - An Huang
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Dong Sun
- Department of Physiology, New York Medical College, Valhalla, New York;
| |
Collapse
|
33
|
Mechanical characteristics of the pulmonary artery in beagle dogs with hepatopulmonary syndrome and portopulmonary hypertension. Biomed Rep 2015; 4:51-54. [PMID: 26870333 DOI: 10.3892/br.2015.526] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/10/2015] [Indexed: 02/07/2023] Open
Abstract
The continuous changes in pulmonary hemodynamic properties in hepatopulmonary syndrome (HPS) and portopulmonary hypertension (PoPH) have not been fully characterized in large animal models of HPS and PoPH. Beagle dog models of HPS and PoPH were induced by chronic common bile duct ligation and Sephadex microspheres, respectively. The model was validated by catheter examination and pathological analyses, and the hemodynamic characteristics of the models were observed. The results revealed that the cross-sectional area of the blood vessel was significantly increased in HPS models, but it was significantly decreased in the PoPH models. Furthermore, the resistance of pulmonary circulation was elevated in models of HPS, but it was decreased in models of PoPH. The present findings renew the traditional view that pulmonary hypertension is due to the enhanced peripheral resistance.
Collapse
|
34
|
Jin Y, Wang W, Chai S, Liu J, Yang T, Wang J. Wnt5a attenuates hypoxia-induced pulmonary arteriolar remodeling and right ventricular hypertrophy in mice. Exp Biol Med (Maywood) 2015; 240:1742-51. [PMID: 25956683 DOI: 10.1177/1535370215584889] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/24/2015] [Indexed: 12/14/2022] Open
Abstract
Hypoxic pulmonary hypertension (HPH), which is characterized by pulmonary arteriolar remodeling and right ventricular hypertrophy, is still a life-threatening disease with the current treatment strategies. The underlying molecular mechanisms of HPH remain unclear. Our previously published study showed that Wnt5a, one of the ligands in the Wnt family, was critically involved in the inhibition of hypoxia-induced pulmonary arterial smooth muscle cell proliferation by downregulation of β-catenin/cyclin D1 in vitro. In this study, we investigated the possible functions and mechanisms of Wnt5a in HPH in vivo. Recombinant mouse Wnt5a (rmWnt5a) or phosphate buffered saline (PBS) was administered to male C57/BL6 mice weekly from the first day to the end of the two or four weeks after exposed to hypoxia (10% O2). Hypoxia-induced pulmonary hypertension was associated with a marked increase in β-catenin/cyclin D1 expression in lungs. Right ventricular systolic pressure and right ventricular hypertrophy index were reduced in animals treated with rmWnt5a compared with PBS. Histology showed less pulmonary vascular remodeling and right ventricular hypertrophy in the group treated with rmWnt5a than with PBS. Treatment with rmWnt5a resulted in a concomitant reduction in β-catenin/cyclin D1 levels in lungs. These data demonstrate that Wnt5a exerts its beneficial effects on HPH by regulating pulmonary vascular remodeling and right ventricular hypertrophy in a manner that is associated with reduction in β-catenin/cyclin D1 signaling. A therapy targeting the β-catenin/cyclin D1 signaling pathway might be a potential strategy for HPH treatment.
Collapse
Affiliation(s)
- Yuling Jin
- Department of Physiology, Capital Medical University, Beijing 100069, P.R. China
| | - Wang Wang
- Department of Physiology, Capital Medical University, Beijing 100069, P.R. China
| | - Sanbao Chai
- Department of Physiology, Capital Medical University, Beijing 100069, P.R. China
| | - Jie Liu
- Department of Physiology, Capital Medical University, Beijing 100069, P.R. China
| | - Ting Yang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100016, P.R. China
| | - Jun Wang
- Department of Physiology, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
35
|
Aguero J, Ishikawa K, Fish KM, Hammoudi N, Hadri L, Garcia-Alvarez A, Ibanez B, Fuster V, Hajjar RJ, Leopold JA. Combination proximal pulmonary artery coiling and distal embolization induces chronic elevations in pulmonary artery pressure in Swine. PLoS One 2015; 10:e0124526. [PMID: 25923775 PMCID: PMC4414513 DOI: 10.1371/journal.pone.0124526] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 03/15/2015] [Indexed: 11/18/2022] Open
Abstract
Pulmonary hypertension (PH) is associated with aberrant vascular remodeling and right ventricular (RV) dysfunction that contribute to early mortality. Large animal models that recapitulate human PH are essential for mechanistic studies and evaluating novel therapies; however, these models are not readily accessible to the field owing to the need for advanced surgical techniques or hypoxia. In this study, we present a novel swine model that develops cardiopulmonary hemodynamics and structural changes characteristic of chronic PH. This percutaneous model was created in swine (n=6) by combining distal embolization of dextran beads with selective coiling of the lobar pulmonary arteries (2 procedures per lung over 4 weeks). As controls, findings from this model were compared with those from a standard weekly distal embolization model (n=6) and sham animals (n=4). Survival with the combined embolization model was 100%. At 8 weeks after the index procedure, combined embolization procedure animals had increased mean pulmonary artery pressure (mPA) and pulmonary vascular resistance (PVR) compared to the controls with no effect on left heart or systemic pressures. RV remodeling and RV dysfunction were also present with a decrease in the RV ejection fraction, increase in the myocardial performance index, impaired longitudinal function, as well as cardiomyocyte hypertrophy, and interstitial fibrosis, which were not present in the controls. Pulmonary vascular remodeling occurred in both embolization models, although only the combination embolization model had a decrease in pulmonary capacitance. Taken together, these cardiopulmonary hemodynamic and structural findings identify the novel combination embolization swine model as a valuable tool for future studies of chronic PH.
Collapse
Affiliation(s)
- Jaume Aguero
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)- Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain
- * E-mail:
| | - Kiyotake Ishikawa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kenneth M. Fish
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Nadjib Hammoudi
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Lahouaria Hadri
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ana Garcia-Alvarez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)- Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)- Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)- Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Roger J. Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Jane A. Leopold
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
36
|
Sanon VP, Sawaki D, Mjaatvedt CH, Jourdan‐Le Saux C. Myocardial Tissue Caveolae. Compr Physiol 2015; 5:871-86. [DOI: 10.1002/cphy.c140050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
37
|
Wilkins MR, Ghofrani HA, Weissmann N, Aldashev A, Zhao L. Pathophysiology and Treatment of High-Altitude Pulmonary Vascular Disease. Circulation 2015; 131:582-90. [DOI: 10.1161/circulationaha.114.006977] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Martin R. Wilkins
- From Experimental Medicine, Imperial College London, Hammersmith Hospital, United Kingdom (M.R.W., H.-A.G., L.Z.); Excellence Cluster Cardio-Pulmonary System, Universities of Giessen, Germany (M.R.W., H.-A.G., N.W., L.Z.); University of Giessen Marburg Lung Center, Justus-Liebig-University, Germany (M.R.W., H.-A.G., N.W., L.Z.); Kerckhoff Clinic, Bad Nauheim, Germany (H.-A.G.); Institute of Molecular Biology and Medicine, Bishkek, Kyrgyzstan (A.A.)
| | - Hossein-Ardeschir Ghofrani
- From Experimental Medicine, Imperial College London, Hammersmith Hospital, United Kingdom (M.R.W., H.-A.G., L.Z.); Excellence Cluster Cardio-Pulmonary System, Universities of Giessen, Germany (M.R.W., H.-A.G., N.W., L.Z.); University of Giessen Marburg Lung Center, Justus-Liebig-University, Germany (M.R.W., H.-A.G., N.W., L.Z.); Kerckhoff Clinic, Bad Nauheim, Germany (H.-A.G.); Institute of Molecular Biology and Medicine, Bishkek, Kyrgyzstan (A.A.)
| | - Norbert Weissmann
- From Experimental Medicine, Imperial College London, Hammersmith Hospital, United Kingdom (M.R.W., H.-A.G., L.Z.); Excellence Cluster Cardio-Pulmonary System, Universities of Giessen, Germany (M.R.W., H.-A.G., N.W., L.Z.); University of Giessen Marburg Lung Center, Justus-Liebig-University, Germany (M.R.W., H.-A.G., N.W., L.Z.); Kerckhoff Clinic, Bad Nauheim, Germany (H.-A.G.); Institute of Molecular Biology and Medicine, Bishkek, Kyrgyzstan (A.A.)
| | - Almaz Aldashev
- From Experimental Medicine, Imperial College London, Hammersmith Hospital, United Kingdom (M.R.W., H.-A.G., L.Z.); Excellence Cluster Cardio-Pulmonary System, Universities of Giessen, Germany (M.R.W., H.-A.G., N.W., L.Z.); University of Giessen Marburg Lung Center, Justus-Liebig-University, Germany (M.R.W., H.-A.G., N.W., L.Z.); Kerckhoff Clinic, Bad Nauheim, Germany (H.-A.G.); Institute of Molecular Biology and Medicine, Bishkek, Kyrgyzstan (A.A.)
| | - Lan Zhao
- From Experimental Medicine, Imperial College London, Hammersmith Hospital, United Kingdom (M.R.W., H.-A.G., L.Z.); Excellence Cluster Cardio-Pulmonary System, Universities of Giessen, Germany (M.R.W., H.-A.G., N.W., L.Z.); University of Giessen Marburg Lung Center, Justus-Liebig-University, Germany (M.R.W., H.-A.G., N.W., L.Z.); Kerckhoff Clinic, Bad Nauheim, Germany (H.-A.G.); Institute of Molecular Biology and Medicine, Bishkek, Kyrgyzstan (A.A.)
| |
Collapse
|
38
|
Tan Y, Tseng PO, Wang D, Zhang H, Hunter K, Hertzberg J, Stenmark KR, Tan W. Stiffening-induced high pulsatility flow activates endothelial inflammation via a TLR2/NF-κB pathway. PLoS One 2014; 9:e102195. [PMID: 25029271 PMCID: PMC4100881 DOI: 10.1371/journal.pone.0102195] [Citation(s) in RCA: 36] [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: 09/21/2013] [Accepted: 06/16/2014] [Indexed: 12/25/2022] Open
Abstract
Stiffening of large arteries is increasingly used as an independent predictor of risk and therapeutic outcome for small artery dysfunction in many diseases including pulmonary hypertension. The molecular mechanisms mediating downstream vascular cell responses to large artery stiffening remain unclear. We hypothesize that high pulsatility flow, induced by large artery stiffening, causes inflammatory responses in downstream pulmonary artery endothelial cells (PAECs) through toll-like receptor (TLR) pathways. To recapitulate the stiffening effect of large pulmonary arteries that occurs in pulmonary hypertension, ultrathin silicone tubes of variable mechanical stiffness were formulated and were placed in a flow circulatory system. These tubes modulated the simulated cardiac output into pulsatile flows with different pulsatility indices, 0.5 (normal) or 1.5 (high). PAECs placed downstream of the tubes were evaluated for their expression of proinflammatory molecules (ICAM-1, VCAM-1, E-selectin and MCP-1), TLR receptors and intracellular NF-κB following flow exposure. Results showed that compared to flow with normal pulsatility, high pulsatility flow induced proinflammatory responses in PAECs, enhanced TLR2 expression but not TLR4, and caused NF-κB activation. Pharmacologic (OxPAPC) and siRNA inhibition of TLR2 attenuated high pulsatility flow-induced pro-inflammatory responses and NF-κB activation in PAECs. We also observed that PAECs isolated from small pulmonary arteries of hypertensive animals exhibiting proximal vascular stiffening demonstrated a durable ex-vivo proinflammatory phenotype (increased TLR2, TLR4 and MCP-1 expression). Intralobar PAECs isolated from vessels of IPAH patients also showed increased TLR2. In conclusion, this study demonstrates for the first time that TLR2/NF-κB signaling mediates endothelial inflammation under high pulsatility flow caused by upstream stiffening, but the role of TLR4 in flow pulsatility-mediated endothelial mechanotransduction remains unclear.
Collapse
Affiliation(s)
- Yan Tan
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, Colorado, United States of America
- Department of Pediatrics, University of Colorado at Denver, Aurora, Colorado, United States of America
| | - Pi-Ou Tseng
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, Colorado, United States of America
| | - Daren Wang
- Department of Pediatrics, University of Colorado at Denver, Aurora, Colorado, United States of America
| | - Hui Zhang
- Department of Pediatrics, University of Colorado at Denver, Aurora, Colorado, United States of America
| | - Kendall Hunter
- Department of Pediatrics, University of Colorado at Denver, Aurora, Colorado, United States of America
| | - Jean Hertzberg
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, Colorado, United States of America
| | - Kurt R. Stenmark
- Department of Pediatrics, University of Colorado at Denver, Aurora, Colorado, United States of America
| | - Wei Tan
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, Colorado, United States of America
| |
Collapse
|
39
|
Ciuclan L, Sheppard K, Dong L, Sutton D, Duggan N, Hussey M, Simmons J, Morrell NW, Jarai G, Edwards M, Dubois G, Thomas M, Van Heeke G, England K. Treatment with anti-gremlin 1 antibody ameliorates chronic hypoxia/SU5416-induced pulmonary arterial hypertension in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 183:1461-73. [PMID: 24160323 DOI: 10.1016/j.ajpath.2013.07.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 06/20/2013] [Accepted: 07/12/2013] [Indexed: 01/05/2023]
Abstract
The expression of the bone morphogenetic protein antagonist, Gremlin 1, was recently shown to be increased in the lungs of pulmonary arterial hypertension patients, and in response to hypoxia. Gremlin 1 released from the vascular endothelium may inhibit endogenous bone morphogenetic protein signaling and contribute to the development of pulmonary arterial hypertension. Here, we investigate the impact of Gremlin 1 inhibition in disease after exposure to chronic hypoxia/SU5416 in mice. We investigated the effects of an anti-Gremlin 1 monoclonal antibody in the chronic hypoxia/SU5416 murine model of pulmonary arterial hypertension. Chronic hypoxic/SU5416 exposure of mice induced upregulation of Gremlin 1 mRNA in lung and right ventricle tissue compared with normoxic controls. Prophylactic treatment with an anti-Gremlin 1 neutralizing mAb reduced the hypoxic/SU5416-dependent increase in pulmonary vascular remodeling and right ventricular hypertrophy. Importantly, therapeutic treatment with an anti-Gremlin 1 antibody also reduced pulmonary vascular remodeling and right ventricular hypertrophy indicating a role for Gremlin 1 in the progression of the disease. We conclude that Gremlin 1 plays a role in the development and progression of pulmonary arterial hypertension in the murine hypoxia/SU5416 model, and that Gremlin 1 is a potential therapeutic target for pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Loredana Ciuclan
- Respiratory Disease Area, Novartis Institutes for BioMedical Research, Horsham, West Sussex, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Chronic overcirculation-induced pulmonary arterial hypertension in aorto-caval shunt. Microvasc Res 2014; 94:73-9. [PMID: 24862700 DOI: 10.1016/j.mvr.2014.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Revised: 05/09/2014] [Accepted: 05/15/2014] [Indexed: 11/20/2022]
Abstract
Pulmonary arterial hypertension is a common complication of congenital heart defects with left-to-right shunts. Current preclinical models do not reproduce clinical characteristics of shunt-related pulmonary hypertension. Aorto-caval shunt was firstly described as a model of right ventricle volume overload. The pathophysiology and the possible determination of pulmonary arterial hypertension of different periods of shunt exposure are still undefined. A method to create standardized, reproducible aorto-caval shunt was developed in growing rats (260±40 g). Three groups of animals were considered: shunt exposure for 10 weeks, shunt exposure for 20 weeks and control (sham laparotomy). Echocardiography and magnetic resonance revealed increased right ventricular end diastolic area in shunt at 10 weeks compared to control. Hemodynamic analysis demonstrated increased right ventricular afterload and increased effective pulmonary arterial elastance (Ea) in shunt at 20 weeks compared to control (1.29±0.20 vs. 0.14±0.06 mmHg/μl, p=0.004). At the same time point, the maximal slope of end-systolic pressure-volume relationship (Ees) decreased (0.5±0.2 mmHg/ml vs. 1.2±0.3, p<0.001). Consequently, right ventricular-arterial coupling was markedly deteriorated with a ≈50% decrease in the ratio of end-systolic to pulmonary artery elastance (Ees/Ea). Finally, left ventricular preload diminished (≈30% decrease in left ventricular end-diastolic volume). Histology demonstrated medial hypertrophy and small artery luminal narrowing. Chronic exposure to aorto-caval shunt is a reliable model to produce right ventricular volume overload and secondary pulmonary arterial hypertension. This model could be an alternative with low mortality and high reproducibility for investigators on the underlying mechanisms of shunt-related pulmonary hypertension.
Collapse
|
41
|
Ibe JCF, Zhou Q, Chen T, Tang H, Yuan JXJ, Raj JU, Zhou G. Adenosine monophosphate-activated protein kinase is required for pulmonary artery smooth muscle cell survival and the development of hypoxic pulmonary hypertension. Am J Respir Cell Mol Biol 2014; 49:609-18. [PMID: 23668615 DOI: 10.1165/rcmb.2012-0446oc] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Human pulmonary artery smooth muscle cells (HPASMCs) express both adenosine monophosphate-activated protein kinase (AMPK) α1 and α2. We investigated the distinct roles of AMPK α1 and α2 in the survival of HPASMCs during hypoxia and hypoxia-induced pulmonary hypertension (PH). The exposure of HPASMCs to hypoxia (3% O2) increased AMPK activation and phosphorylation, and the inhibition of AMPK with Compound C during hypoxia decreased their viability and increased lactate dehydrogenase activity and apoptosis. Although the suppression of either AMPK α1 or α2 expression led to increased cell death, the suppression of AMPK α2 alone increased caspase-3 activity and apoptosis in HPASMCs exposed to hypoxia. It also resulted in the decreased expression of myeloid cell leukemia sequence 1 (MCL-1). The knockdown of MCL-1 or MCL-1 inhibitors increased caspase-3 activity and apoptosis in HPASMCs exposed to hypoxia. On the other hand, the suppression of AMPK α1 expression alone prevented hypoxia-mediated autophagy. The inhibition of autophagy induced cell death in HPASMCs. Our results suggest that AMPK α1 and AMPK α2 play differential roles in the survival of HPASMCs during hypoxia. The activation of AMPK α2 maintains the expression of MCL-1 and prevents apoptosis, whereas the activation of AMPK α1 stimulates autophagy, promoting HPASMC survival. Moreover, treatment with Compound C, which inhibits both isoforms of AMPK, prevented and partly reversed hypoxia-induced PH in mice. Taking these results together, our study suggests that AMPK plays a key role in the pathogenesis of pulmonary arterial hypertension, and AMPK may represent a novel therapeutic target for the treatment of pulmonary arterial hypertension.
Collapse
|
42
|
Tewari SG, Bugenhagen SM, Wang Z, Schreier DA, Carlson BE, Chesler NC, Beard DA. Analysis of cardiovascular dynamics in pulmonary hypertensive C57BL6/J mice. Front Physiol 2013; 4:355. [PMID: 24376421 PMCID: PMC3858724 DOI: 10.3389/fphys.2013.00355] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 11/20/2013] [Indexed: 12/03/2022] Open
Abstract
A computer model was used to analyze data on cardiac and vascular mechanics from C57BL6/J mice exposed to 0 (n = 4), 14 (n = 6), 21 (n = 8) and 28 (n = 7) days of chronic hypoxia and treatment with the VEGF receptor inhibitor SUGEN (HySu) to induce pulmonary hypertension. Data on right ventricular pressure and volume, and systemic arterial pressure obtained before, during, and after inferior vena cava occlusion were analyzed using a mathematical model of realistic ventricular mechanics coupled with a simple model of the pulmonary and systemic vascular systems. The model invokes a total of 26 adjustable parameters, which were estimated based on least-squares fitting of the data. Of the 26 adjustable parameters, 14 were set to globally constant values for the entire data set. It was necessary to adjust the remaining 12 parameters to match data from all experimental groups. Of these 12 individually adjusted parameters, three parameters representing pulmonary vascular resistance, pulmonary arterial elastance, and pulmonary arterial narrowing were found to significantly change in HySu-induced remodeling. Model analysis shows a monotonic change in these parameters as disease progressed, with approximately 130% increase in pulmonary resistance, 70% decrease in unstressed pulmonary arterial volume, and 110% increase in pulmonary arterial elastance in the 28-day group compared to the control group. These changes are consistent with prior experimental measurements. Furthermore, the 28-day data could be explained only after increasing the passive elastance of the right free wall compared to the value used for the other data sets, which is likely a consequence of the increased RV collagen accumulation found experimentally. These findings may indicate a compensatory remodeling followed by pathological remodeling of the right ventricle in HySu-induced pulmonary hypertension.
Collapse
Affiliation(s)
- Shivendra G Tewari
- Biotechnology and Bioengineering Center, Medical College of Wisconsin Milwaukee, WI, USA
| | - Scott M Bugenhagen
- Biotechnology and Bioengineering Center, Medical College of Wisconsin Milwaukee, WI, USA ; Department of Physiology, Medical College of Wisconsin Milwaukee, WI, USA
| | - Zhijie Wang
- Department of Biomedical Engineering, University of Wisconsin-Madison Madison, WI, USA
| | - David A Schreier
- Department of Biomedical Engineering, University of Wisconsin-Madison Madison, WI, USA
| | - Brian E Carlson
- Biotechnology and Bioengineering Center, Medical College of Wisconsin Milwaukee, WI, USA ; Department of Physiology, Medical College of Wisconsin Milwaukee, WI, USA
| | - Naomi C Chesler
- Department of Biomedical Engineering, University of Wisconsin-Madison Madison, WI, USA
| | - Daniel A Beard
- Biotechnology and Bioengineering Center, Medical College of Wisconsin Milwaukee, WI, USA ; Department of Physiology, Medical College of Wisconsin Milwaukee, WI, USA
| |
Collapse
|
43
|
Bakhshi FR, Mao M, Shajahan AN, Piegeler T, Chen Z, Chernaya O, Sharma T, Elliott WM, Szulcek R, Bogaard HJ, Comhair S, Erzurum S, van Nieuw Amerongen GP, Bonini MG, Minshall RD. Nitrosation-dependent caveolin 1 phosphorylation, ubiquitination, and degradation and its association with idiopathic pulmonary arterial hypertension. Pulm Circ 2013; 3:816-30. [PMID: 25006397 PMCID: PMC4070841 DOI: 10.1086/674753] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 09/18/2013] [Indexed: 01/15/2023] Open
Abstract
In the present study, we tested the hypothesis that chronic inflammation and oxidative/nitrosative stress induce caveolin 1 (Cav-1) degradation, providing an underlying mechanism of endothelial cell activation/dysfunction and pulmonary vascular remodeling in patients with idiopathic pulmonary arterial hypertension (IPAH). We observed reduced Cav-1 protein despite increased Cav-1 messenger RNA expression and also endothelial nitric oxide synthase (eNOS) hyperphosphorylation in human pulmonary artery endothelial cells (PAECs) from patients with IPAH. In control human lung endothelial cell cultures, tumor necrosis factor α-induced nitric oxide (NO) production and S-nitrosation (SNO) of Cav-1 Cys-156 were associated with Src displacement and activation, Cav-1 Tyr-14 phosphorylation, and destabilization of Cav-1 oligomers within 5 minutes that could be blocked by eNOS or Src inhibition. Prolonged stimulation (72 hours) with NO donor DETANONOate reduced oligomerized and total Cav-1 levels by 40%-80%, similar to that observed in IPAH patient-derived PAECs. NO donor stimulation of endothelial cells for >72 hours, which was associated with sustained Src activation and Cav-1 phosphorylation, ubiquitination, and degradation, was blocked by NOS inhibitor L-NAME, Src inhibitor PP2, and proteosomal inhibitor MG132. Thus, chronic inflammation, sustained eNOS and Src signaling, and Cav-1 degradation may be important causal factors in the development of IPAH by promoting PAEC dysfunction/activation via sustained oxidative/nitrosative stress.
Collapse
Affiliation(s)
- Farnaz R. Bakhshi
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Mao Mao
- Department of Medicine, Section of Cardiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Ayesha N. Shajahan
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Tobias Piegeler
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Zhenlong Chen
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Olga Chernaya
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Tiffany Sharma
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - W. Mark Elliott
- Pulmonary Division, James Hogg Research Centre Biobank, University of British Columbia, Vancouver, Canada
| | - Robert Szulcek
- Department of Physiology, Institute for Cardiovascular Research, Vrije Universiteit (VU) University Medical Center, Amsterdam, The Netherlands
- Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Harm Jan Bogaard
- Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Suzy Comhair
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Serpil Erzurum
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Geerten P. van Nieuw Amerongen
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Physiology, Institute for Cardiovascular Research, Vrije Universiteit (VU) University Medical Center, Amsterdam, The Netherlands
| | - Marcelo G. Bonini
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Medicine, Section of Cardiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Richard D. Minshall
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
44
|
Original rat model of high kinetic unilateral pulmonary hypertension surgically induced by combined surgery. J Thorac Cardiovasc Surg 2013; 146:1220-1226.e1. [DOI: 10.1016/j.jtcvs.2013.01.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 12/09/2012] [Accepted: 01/11/2013] [Indexed: 11/19/2022]
|
45
|
Yue J, Guan J, Wang X, Zhang L, Yang Z, Ao Q, Deng Y, Zhu P, Wang G. MicroRNA-206 is involved in hypoxia-induced pulmonary hypertension through targeting of the HIF-1α/Fhl-1 pathway. J Transl Med 2013; 93:748-59. [PMID: 23628900 DOI: 10.1038/labinvest.2013.63] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hypoxia-induced pulmonary hypertension (PH), which is characterized by vasoconstriction and subsequent structural remodeling of blood vessels, is an important event in chronic obstructive pulmonary disease patients and in people living at high altitudes. Hypoxia-inducible factor-1α (HIF-1α) and its regulator four-and-a-half LIM (Lin-11, Isl-1 and Mec-3) domain 1 (Fhl-1) have important roles in hypoxia-induced PH. MicroRNA-206 (miR-206) is critical for myogenesis and related diseases; however, the role of miR-206 in hypoxia-induced PH is unknown. miR-206 expression was evaluated in a hypoxic rat model and in cultured hypoxic pulmonary artery smooth muscle cells (PASMCs) using real-time quantitative PCR (RT-qPCR). HIF-1α and Fhl-1 expression were evaluated using RT-qPCR, western blotting, immunohistochemistry and immunofluorescence. The function of miR-206 was assessed by transfecting miR-206 mimics and inhibitors. Dual-luciferase reporter gene assays and western blotting were performed to validate the target genes of miR-206. siRNA targeted against Fhl-1 was used to investigate the effect of Fhl-1 on miR-206. Flow cytometry was used to detect the cell cycle phase distribution in each group of PASMCs. Significant downregulation of miR-206 in hypoxic lung tissue and PASMCs was identified, whereas HIF-1α and Fhl-1 were upregulated in these samples. The expression of miR-206 in the serum was different from that in the lung tissue. Transfection of pre-miR miR-206 in hypoxic conditions led to increased expression of HIF-1α and Fhl-1 rather than abolishing hypoxia-induced HIF-1α and Fhl-1, as was expected, and promoted the entry of cells into the S phase and enhanced PASMC proliferation. Fhl-1-targeted siRNA in PASMC prevented cell proliferation and led to an increased proportion of cells in the G1 phase without altering miR-206 expression. Bioinformatic analysis and dual-luciferase reporter gene assays revealed direct evidence for miR-206 targeting of HIF-1α. In conclusion, hypoxia-induced downregulation of miR-206 promotes PH by targeting the HIF-1α/Fhl-1 pathway in PASMCs. miR-206 could be a triggering factor of early stage of hypoxia-induced PH.
Collapse
Affiliation(s)
- Junqiu Yue
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Plakkal N, Soraisham AS, Trevenen C, Freiheit EA, Sauve R. Histological chorioamnionitis and bronchopulmonary dysplasia: a retrospective cohort study. J Perinatol 2013; 33:441-5. [PMID: 23238570 DOI: 10.1038/jp.2012.154] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To examine the association between histological chorioamnionitis (HC) with or without fetal inflammatory response (FIR) and bronchopulmonary dysplasia (BPD) in preterm infants. STUDY DESIGN We conducted a retrospective cohort study of infants born at <29 weeks gestation admitted to the neonatal intensive care unit from 2000 to 2006, who had placental histology. We compared the incidence of BPD among three groups: No HC group, HC without FIR group and HC with FIR group. The multivariable model based on generalized estimating equation was fitted to estimate the adjusted risk ratios (aRR) and 95% confidence intervals (CIs) for BPD and combined outcome of BPD or death. RESULT Of 529 infants, 84 (16%) had HC without FIR, 186 (35%) had HC with FIR and 259 (49%) had no HC. Compared with the no HC group, HC with and without FIR group infants were of lower gestational age and singleton births. Multivariable modeling based on generalized estimating equation revealed that HC with FIR is associated with decreased risk of both BPD (aRR 0.88, 95% CI 0.81 to 0.95) and the combined outcome of BPD or death (aRR 0.91, 95% CI 0.86 to 0.97). HC without FIR showed a trend toward reduction in BPD (aRR 0.93, 95% CI 0.86 to 1.00). CONCLUSIONS HC with FIR is associated with decreased risk of both BPD and the combined outcome of BPD or death in preterm infants.
Collapse
Affiliation(s)
- N Plakkal
- Department of Pediatrics and Alberta Children's Hospital Research Institute for Child and Maternal Health, University of Calgary, Calgary, AT, Canada
| | | | | | | | | |
Collapse
|
47
|
Zuo X, Zong F, Wang H, Wang Q, Xie W, Wang H. Iptakalim, a novel ATP-sensitive potassium channel opener, inhibits pulmonary arterial smooth muscle cell proliferation by downregulation of PKC-α. J Biomed Res 2013; 25:392-401. [PMID: 23554716 PMCID: PMC3596718 DOI: 10.1016/s1674-8301(11)60052-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 03/25/2011] [Accepted: 05/20/2011] [Indexed: 12/17/2022] Open
Abstract
Iptakalim is a new ATP-sensitive potassium (KATP) channel opener, and it inhibits the proliferation of pulmonary arterial smooth muscle cells (PASMCs) and pulmonary vascular remodeling. However, the underlying mechanism remains unclear. In the present study, we found that iptakalim significantly decreased pulmonary artery pressure, inhibited pulmonary ariery remodeling and PKC-α overexpression in chronic hypoxia in a rat pulmonary hypertension model. Iptakalim reduced hypoxia-induced expression of PKC-α, and abolished the effect of hypoxia on PASMC proliferation significantly in a dose-dependent manner in vitro. Moreover, these effects were abolished by glibenclamide, a selective KATP channel antagonist. These results indicate that iptakalim inhibits PASMC proliferation and pulmonary vascular remodeling induced by hypoxia through downregulating the expression of PKC-α. Iptakalim can serve as a novel promising treatment for hypoxic pulmonary hypertension.
Collapse
Affiliation(s)
- Xiangrong Zuo
- Department of Respiratory Medicine; ; Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | | | | | | | | | | |
Collapse
|
48
|
Wei L, Zhu W, Xia L, Yang Y, Liu H, Shen J, Zhu J, Xu Y, Yang Z, Wang C. Therapeutic effect of eNOS-transfected endothelial progenitor cells on hemodynamic pulmonary arterial hypertension. Hypertens Res 2013; 36:414-21. [PMID: 23446773 DOI: 10.1038/hr.2012.217] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hemodynamic pulmonary arterial hypertension (HPAH) is a common symptom in congenital heart disease (CHD) patients with a left-to-right shunt. Endothelial NO synthase (eNOS) and endothelial-like progenitor cells result in significant improvement of right ventricular systolic pressure in established pulmonary arterial hypertension (PAH) models. We hypothesized that bone marrow (BM)-derived endothelial progenitor cells (EPCs) and eNOS would prevent HPAH in a newly established rat model. The heNOS gene was cloned into a PSUCMV vector, and a high-titer adenovirus was generated. Mononuclear cells (MNCs) from rat BM were differentiated into EPCs by treatment with various cytokines, and a high purity of EPCs (>70%) was confirmed using the markers DiI ac-LDL, UEA-1, vWF and Flk-1. An ideal rat HPAH model was successfully established based on right lung lobectomy, and was confirmed by pressure measurement and histological staining. heNOS was successfully transfected into EPCs, which were then transplanted into HPAH rats. Two weeks after transplantation, the systolic pulmonary arterial blood pressure (sPAP) was significantly reduced by heNOS-EPCs treatment and by transplantation of control EPCs. The high number of muscular pulmonary arteries and the thickness of the muscular coat characteristic of HPAH rats were clearly reversed or even restored to normal levels following transplantation of EPCs, particularly eNOS-EPCs. These findings indicate a critical role of eNOS in HPAH treatment and suggest that eNOS-transfected EPCs may provide an effective strategy for HPAH treatment in CHD patients.
Collapse
Affiliation(s)
- Lai Wei
- Department of Cardiac Surgery, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Voelkel NF, Mizuno S, Bogaard HJ. The role of hypoxia in pulmonary vascular diseases: a perspective. Am J Physiol Lung Cell Mol Physiol 2013; 304:L457-65. [PMID: 23377344 DOI: 10.1152/ajplung.00335.2012] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
From the discovery of hypoxic pulmonary vasoconstriction, responses to hypoxia have been considered as representative for the many alterations in lung vessels that occur in several chronic lung diseases, including pulmonary hypertension, interstitial pulmonary fibrosis, acute respiratory distress syndrome, and chronic obstructive pulmonary disease. An essential part of preclinical research to explain the pathobiology of these diseases has been centered on the exposure of small and large animals to hypoxia. This review aims to summarize pivotal results of clinical and preclinical research on hypoxia, which still have important implications for researchers today.
Collapse
Affiliation(s)
- Norbert F Voelkel
- Victoria Johnson Laboratory for Lung Research, Pulmonary and Critical Care Medicine Division, Virginia Commonwealth University, Richmond, VA, USA
| | | | | |
Collapse
|
50
|
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
|