1
|
Drummond FR, Leite LB, de Miranda DC, Drummond LR, Lavorato VN, Soares LL, Neves CA, Natali AJ. Skeletal muscle dysfunctions in pulmonary arterial hypertension: Effects of aerobic exercise training. Front Physiol 2023; 14:1148146. [PMID: 37035672 PMCID: PMC10076612 DOI: 10.3389/fphys.2023.1148146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Pulmonary arterial hypertension is associated with skeletal muscle myopathy and atrophy and impaired exercise tolerance. Aerobic exercise training has been recommended as a non-pharmacological therapy for deleterious effects imposed by pulmonary arterial hypertension. Aerobic physical training induces skeletal muscle adaptations via reduced inflammation, improved anabolic processes, decreased hypoxia and regulation of mitochondrial function. These benefits improve physical exertion tolerance and quality of life in patients with pulmonary arterial hypertension. However, the mechanisms underlying the therapeutic potential of aerobic exercise to skeletal muscle disfunctions in patients with pulmonary arterial hypertension are not well understood yet. This minireview highlights the pathways involved in skeletal muscle adaptations to aerobic exercise training in patients with pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Filipe Rios Drummond
- Department of General Biology, Laboratory of Structural Biology, Federal University of Viçosa, Viçosa, MG, Brazil
| | - Luciano Bernardes Leite
- Department of Physical Education, Laboratory of Exercise Biology Federal University of Viçosa, Viçosa, MG, Brazil
- *Correspondence: Luciano Bernardes Leite,
| | - Denise Coutinho de Miranda
- Department of Biological Sciences, Laboratory of Cell Signaling, Federal University of Ouro Preto, Viçosa, MG, Brazil
- Department of Physical Education, Governador Ozanam Coelho University Center (UNIFAGOC), Ubá, Brazil
| | - Lucas Rios Drummond
- Department of Physiology and Biophysics, Laboratory of Endocrinology and Metabolism, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Victor Neiva Lavorato
- Department of Physical Education, Governador Ozanam Coelho University Center (UNIFAGOC), Ubá, Brazil
| | - Leôncio Lopes Soares
- Department of Physical Education, Laboratory of Exercise Biology Federal University of Viçosa, Viçosa, MG, Brazil
| | - Clóvis Andrade Neves
- Department of General Biology, Laboratory of Structural Biology, Federal University of Viçosa, Viçosa, MG, Brazil
| | - Antônio José Natali
- Department of Physical Education, Laboratory of Exercise Biology Federal University of Viçosa, Viçosa, MG, Brazil
| |
Collapse
|
2
|
Swinarew AS, Gabor J, Kusz B, Skoczyński S, Raif P, Skoczylas I, Jonas K, Grabka M, Mizia-Szubryt M, Bula K, Stanula A, Mika B, Tkacz E, Paluch J, Gąsior M, Kopeć G, Mizia-Stec K. Exhaled Air Metabolome Analysis for Pulmonary Arterial Hypertension Fingerprints Identification-The Preliminary Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 20:503. [PMID: 36612835 PMCID: PMC9819134 DOI: 10.3390/ijerph20010503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 06/17/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease with a serious prognosis. The aim of this study was to identify biomarkers for PAH in the breath phase and to prepare an automatic classification method to determine the changing metabolome trends and molecular mapping. A group of 37 patients (F/M: 8/29 women, mean age 60.4 ± 10.9 years, BMI 27.6 ± 6.0 kg/m2) with diagnosed PAH were enrolled in the study. The breath phase of all the patients was collected on a highly porous septic material using a special patented holder PL230578, OHIM 002890789-0001. The collected air was then examined with gas chromatography coupled with mass spectrometry (GC/MS). The algorithms of Spectral Clustering, KMeans, DBSCAN, and hierarchical clustering methods were used to perform the cluster analysis. The identification of the changes in the ratio of the whole spectra of biomarkers allowed us to obtain a multidimensional pathway for PAH characteristics and showed the metabolome differences in the four subgroups divided by the cluster analysis. The use of GC/MS, supported with novel porous polymeric materials, for the breath phase analysis seems to be a useful tool in selecting bio-fingerprints in patients with PAH. The four metabolome classes which were obtained constitute novel data in the PAH population.
Collapse
Affiliation(s)
- Andrzej S. Swinarew
- Faculty of Science and Technology, University of Silesia in Katowice, 41-500 Chorzów, Poland
- Department of Swimming and Water Rescue, Institute of Sport Science, The Jerzy Kukuczka Academy of Physical Education, 40-065 Katowice, Poland
| | - Jadwiga Gabor
- Faculty of Science and Technology, University of Silesia in Katowice, 41-500 Chorzów, Poland
| | - Błażej Kusz
- First Department of Cardiology, Faculty of Medicine in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
| | - Szymon Skoczyński
- Department of Pneumonology, Faculty of Medicine in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
| | - Paweł Raif
- Department of Biosensors and Biomedical Signals Processing, Silesian University of Technology, 41-800 Zabrze, Poland
| | - Ilona Skoczylas
- 3rd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-800 Katowice, Poland
| | - Kamil Jonas
- Pulmonary Circulation Centre, Department of Cardiac and Vascular Diseases, Jagiellonian University Medical College, John Paul II Hospital in Krakow, 31-349 Kraków, Poland
| | - Marek Grabka
- First Department of Cardiology, Faculty of Medicine in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
| | - Magdalena Mizia-Szubryt
- First Department of Cardiology, Faculty of Medicine in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
| | - Karolina Bula
- First Department of Cardiology, Faculty of Medicine in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
| | - Arkadiusz Stanula
- Department of Swimming and Water Rescue, Institute of Sport Science, The Jerzy Kukuczka Academy of Physical Education, 40-065 Katowice, Poland
| | - Barbara Mika
- Department of Biosensors and Biomedical Signals Processing, Silesian University of Technology, 41-800 Zabrze, Poland
| | - Ewaryst Tkacz
- Department of Biosensors and Biomedical Signals Processing, Silesian University of Technology, 41-800 Zabrze, Poland
| | - Jarosław Paluch
- Department of ENT, Faculty of Medical Sciences in Katowice, Medical University Silesia, 40-055 Katowice, Poland
| | - Mariusz Gąsior
- 3rd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-800 Katowice, Poland
| | - Grzegorz Kopeć
- Pulmonary Circulation Centre, Department of Cardiac and Vascular Diseases, Jagiellonian University Medical College, John Paul II Hospital in Krakow, 31-349 Kraków, Poland
| | - Katarzyna Mizia-Stec
- First Department of Cardiology, Faculty of Medicine in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
| |
Collapse
|
3
|
Somnarin T, Pobsuk N, Chantakul R, Panklai T, Temkitthawon P, Hannongbua S, Chootip K, Ingkaninan K, Boonyarattanakalin K, Gleeson D, Paul Gleeson M. Computational design, synthesis and biological evaluation of PDE5 inhibitors based on N 2,N 4-diaminoquinazoline and N 2,N 6-diaminopurine scaffolds. Bioorg Med Chem 2022; 76:117092. [PMID: 36450167 DOI: 10.1016/j.bmc.2022.117092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 11/18/2022]
Abstract
We report the synthesis, and characterization of twenty-nine new inhibitors of PDE5. Structure-based design was employed to modify to our previously reported 2,4-diaminoquinazoline series. Modification include scaffold hopping to 2,6-diaminopurine core as well as incorporation of ionizable groups to improve both activity and solubility. The prospective binding mode of the compounds was determined using 3D ligand-based similarity methods to inhibitors of known binding mode, combined with a PDE5 docking and molecular dynamics based-protocol, each of which pointed to the same binding mode. Chemical modifications were then designed to both increase potency and solubility as well as validate the binding mode prediction. Compounds containing a quinazoline core displayed IC50s ranging from 0.10 to 9.39 µM while those consisting of a purine scaffold ranging from 0.29 to 43.16 µM. We identified 25 with a PDE5 IC50 of 0.15 µM, and much improved solubility (1.77 mg/mL) over the starting lead. Furthermore, it was found that the predicted binding mode was consistent with the observed SAR validating our computationally driven approach.
Collapse
Affiliation(s)
- Thanachon Somnarin
- Department of Biomedical Engineering, School of Engineering, King Mongkut's Institute of Technology Ladkrabang, Bangkok 10520, Thailand
| | - Nattakarn Pobsuk
- Department of Chemistry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Ruttanaporn Chantakul
- Center of Excellence in Cannabis Research, Faculty of Pharmaceutical Sciences & Center of Excellence in Innovation in Chemistry, Naresuan University, Phitsanulok 65000, Thailand
| | - Teerapap Panklai
- Center of Excellence in Cannabis Research, Faculty of Pharmaceutical Sciences & Center of Excellence in Innovation in Chemistry, Naresuan University, Phitsanulok 65000, Thailand
| | - Prapapan Temkitthawon
- Center of Excellence in Cannabis Research, Faculty of Pharmaceutical Sciences & Center of Excellence in Innovation in Chemistry, Naresuan University, Phitsanulok 65000, Thailand
| | - Supa Hannongbua
- Department of Chemistry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Krongkarn Chootip
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Kornkanok Ingkaninan
- Center of Excellence in Cannabis Research, Faculty of Pharmaceutical Sciences & Center of Excellence in Innovation in Chemistry, Naresuan University, Phitsanulok 65000, Thailand.
| | - Kanokthip Boonyarattanakalin
- College of Materials Innovation and Technology, King Mongkut's Institute of Technology Ladkrabang, Bangkok 10520, Thailand
| | - Duangkamol Gleeson
- Department of Chemistry & Applied Computational Chemistry Research Unit, School of Science, King Mongkut's Institute of Technology Ladkrabang, Bangkok 10520, Thailand
| | - M Paul Gleeson
- Department of Biomedical Engineering, School of Engineering, King Mongkut's Institute of Technology Ladkrabang, Bangkok 10520, Thailand.
| |
Collapse
|
4
|
Wang Y, Lin L, Li X, Cao J, Wang J, Jing ZC, Li S, Liu H, Wang X, Jin ZY, Wang YN. Native T1 Mapping-Based Radiomics for Noninvasive Prediction of the Therapeutic Effect of Pulmonary Arterial Hypertension. Diagnostics (Basel) 2022; 12:diagnostics12102492. [PMID: 36292180 PMCID: PMC9600513 DOI: 10.3390/diagnostics12102492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Novel markers for predicting the short-term therapeutic effect of pulmonary arterial hypertension (PAH) to assist in the prompt initiation of tailored treatment strategies are greatly needed and highly desirable. The aim of the study was to investigate the role of cardiac magnetic resonance (CMR) native T1 mapping radiomics in predicting the short-term therapeutic effect in PAH patients; (2) Methods: Fifty-five PAH patients who received targeted therapy were retrospectively included. Patients were subdivided into an effective group and an ineffective group by assessing the therapeutic effect after ≥3 months of treatment. All patients underwent CMR examinations prior to the beginning of the therapy. Radiomics features from native T1 mapping images were extracted. A radiomics model was constructed using the support vector machine (SVM) algorithm for predicting the therapeutic effect; (3) Results: The SVM radiomics model revealed favorable performance for predicting the therapeutic effect with areas under the receiver operating characteristic curve of 0.955 in the training cohort and 0.893 in the test cohort, respectively. With the optimal cutoff value, the radiomics model showed accuracies of 0.909 and 0.818 in the training and test cohorts, respectively; (4) Conclusions: The CMR native T1 mapping-based radiomics model holds promise for predicting the therapeutic effect in PAH patients.
Collapse
Affiliation(s)
- Yue Wang
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, No. 1, Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Lu Lin
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, No. 1, Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Xiao Li
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, No. 1, Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Jian Cao
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, No. 1, Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Jian Wang
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, No. 1, Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Zhi-Cheng Jing
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, No. 1, Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Sen Li
- Department of Research & Development, Yizhun Medical AI Co., Ltd., 12th Floor 12, Block A, Beihang Zhizhen Building, No. 7 Zhichun Road, Haidian District, Beijing 100088, China
| | - Hao Liu
- Department of Research & Development, Yizhun Medical AI Co., Ltd., 12th Floor 12, Block A, Beihang Zhizhen Building, No. 7 Zhichun Road, Haidian District, Beijing 100088, China
| | - Xin Wang
- Department of Research & Development, Yizhun Medical AI Co., Ltd., 12th Floor 12, Block A, Beihang Zhizhen Building, No. 7 Zhichun Road, Haidian District, Beijing 100088, China
| | - Zheng-Yu Jin
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, No. 1, Shuaifuyuan, Dongcheng District, Beijing 100730, China
- Correspondence: (Y.-N.W.); (Z.-Y.J.)
| | - Yi-Ning Wang
- Department of Radiology, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, No. 1, Shuaifuyuan, Dongcheng District, Beijing 100730, China
- Correspondence: (Y.-N.W.); (Z.-Y.J.)
| |
Collapse
|
5
|
Rhodes CJ, Wharton J, Swietlik EM, Harbaum L, Girerd B, Coghlan JG, Lordan J, Church C, Pepke-Zaba J, Toshner M, Wort SJ, Kiely DG, Condliffe R, Lawrie A, Gräf S, Montani D, Boucly A, Sitbon O, Humbert M, Howard LS, Morrell NW, Wilkins MR. Using the Plasma Proteome for Risk Stratifying Patients with Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2022; 205:1102-1111. [PMID: 35081018 PMCID: PMC9851485 DOI: 10.1164/rccm.202105-1118oc] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Rationale: NT-proBNP (N-terminal pro-brain natriuretic peptide), a biomarker of cardiac origin, is used to risk stratify patients with pulmonary arterial hypertension (PAH). Its limitations include poor sensitivity to early vascular pathology. Other biomarkers of vascular or systemic origin may also be useful in the management of PAH. Objectives: Identify prognostic proteins in PAH that complement NT-proBNP and clinical risk scores. Methods: An aptamer-based assay (SomaScan version 4) targeting 4,152 proteins was used to measure plasma proteins in patients with idiopathic, heritable, or drug-induced PAH from the UK National Cohort of PAH (n = 357) and the French EFORT (Evaluation of Prognostic Factors and Therapeutic Targets in PAH) study (n = 79). Prognostic proteins were identified in discovery-replication analyses of UK samples. Proteins independent of 6-minute-walk distance and NT-proBNP entered least absolute shrinkage and selection operator modeling, and the best combination in a single score was evaluated against clinical targets in EFORT. Measurements and Main Results: Thirty-one proteins robustly informed prognosis independent of NT-proBNP and 6-minute-walk distance in the UK cohort. A weighted combination score of six proteins was validated at baseline (5-yr mortality; area under the curve [AUC], 0.73; 95% confidence interval [CI], 0.63-0.85) and follow-up in EFORT (AUC, 0.84; 95% CI, 0.75-0.94; P = 9.96 × 10-6). The protein score risk stratified patients independent of established clinical targets and risk equations. The addition of the six-protein model score to NT-proBNP improved prediction of 5-year outcomes from AUC 0.762 (0.702-0.821) to 0.818 (0.767-0.869) by receiver operating characteristic analysis (P = 0.00426 for difference in AUC) in the UK replication and French samples combined. Conclusions: The plasma proteome informs prognosis beyond established factors in PAH and may provide a more sensitive measure of therapeutic response.
Collapse
Affiliation(s)
- Christopher J Rhodes
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - John Wharton
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Emilia M Swietlik
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Lars Harbaum
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Barbara Girerd
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - J Gerry Coghlan
- Department of Cardiology, Royal Free Campus, University College London, London, United Kingdom
| | - James Lordan
- University of Newcastle upon Tyne, Newcastle upon Tyne, United Kingdom
| | - Colin Church
- University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Joanna Pepke-Zaba
- Pulmonary Vascular Disease Unit, Royal Papworth Hospital, Cambridge, United Kingdom
| | - Mark Toshner
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Stephen J Wort
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - David G Kiely
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom.,Sheffield Pulmonary Vascular Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom; and
| | - Robin Condliffe
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom.,Sheffield Pulmonary Vascular Unit, Royal Hallamshire Hospital, Sheffield, United Kingdom; and
| | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,BioResource for Translational Research, National Institute for Health Research Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - David Montani
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Athénaïs Boucly
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Olivier Sitbon
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Marc Humbert
- Université Paris-Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Luke S Howard
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | |
Collapse
|
6
|
Kv7 Channels in Cyclic-Nucleotide Dependent Relaxation of Rat Intra-Pulmonary Artery. Biomolecules 2022; 12:biom12030429. [PMID: 35327621 PMCID: PMC8946781 DOI: 10.3390/biom12030429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/22/2022] Open
Abstract
Pulmonary hypertension is treated with drugs that stimulate cGMP or cAMP signalling. Both nucleotides can activate Kv7 channels, leading to smooth muscle hyperpolarisation, reduced Ca2+ influx and relaxation. Kv7 activation by cGMP contributes to the pulmonary vasodilator action of nitric oxide, but its contribution when dilation is evoked by the atrial natriuretic peptide (ANP) sensitive guanylate cyclase, or cAMP, is unknown. Small vessel myography was used to investigate the ability of Kv7 channel blockers to interfere with pulmonary artery relaxation when cyclic nucleotide pathways were stimulated in different ways. The pan-Kv7 blockers, linopirdine and XE991, caused substantial inhibition of relaxation evoked by NO donors and ANP, as well as endothelium-dependent dilators, the guanylate cyclase stimulator, riociguat, and the phosphodiesterase-5 inhibitor, sildenafil. Maximum relaxation was reduced without a change in sensitivity. The blockers had relatively little effect on cAMP-mediated relaxation evoked by forskolin, isoprenaline or treprostinil. The Kv7.1-selective blocker, HMR1556, had no effect on cGMP or cAMP-dependent relaxation. Western blot analysis demonstrated the presence of Kv7.1 and Kv7.4 proteins, while selective activators of Kv7.1 and Kv7.4 homomeric channels, but not Kv7.5, caused pulmonary artery relaxation. It is concluded that Kv7.4 channels contribute to endothelium-dependent dilation and the effects of drugs that act by stimulating cGMP, but not cAMP, signalling.
Collapse
|
7
|
Ferrari TCA, Albricker ACL, Gonçalves IM, Freire CMV. Schistosome-Associated Pulmonary Arterial Hypertension: A Review Emphasizing Pathogenesis. Front Cardiovasc Med 2021; 8:724254. [PMID: 34676250 PMCID: PMC8523797 DOI: 10.3389/fcvm.2021.724254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/13/2021] [Indexed: 12/21/2022] Open
Abstract
Schistosomiasis, especially due to Schistosoma mansoni, is a well-recognized cause of pulmonary arterial hypertension (PAH). The high prevalence of this helminthiasis makes schistosome-related PAH (Sch-PAH) one of the most common causes of this disorder worldwide. The pathogenic mechanisms underlying Sch-PAH remain largely unknown. Available evidence suggests that schistosome eggs reach the lung via portocaval shunts formed as a consequence of portal hypertension due to hepatosplenic schistosomiasis. Once deposited into the lungs, the eggs elicit an immune response resulting in periovular granuloma formation. Immune mediators drive transforming growth factor-β (TGF-β) release, which gives rise to pulmonary vascular inflammation with subsequent remodeling and development of angiomatoid and plexiform lesions. These mechanisms elicited by the eggs seem to become autonomous and the vascular lesions progress independently of the antigen. Portopulmonary hypertension, which pathogenesis is still uncertain, may also play a role in the genesis of Sch-PAH. Recently, there have been substantial advances in the diagnosis and treatment of PAH, but it remains a difficult condition to recognize and manage, and patients still die prematurely from right-heart failure. Echocardiography is used for screening, and the formal diagnosis requires right-heart catheterization. The experience in treating Sch-PAH is largely limited to the phosphodiesterase type 5 inhibitors, with evidence suggesting that these vasodilators improve symptoms and may also improve survival. Considering the great deal of uncertainty about Sch-PAH pathogenesis, course, and treatment, the aim of this review is to summarize current knowledge on this condition emphasizing its pathogenesis.
Collapse
Affiliation(s)
- Teresa Cristina Abreu Ferrari
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Cristina Lopes Albricker
- Programa de Pós-Graduação em Ciências Aplicadas à Saúde do Adulto, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ina Morais Gonçalves
- Graduação em Medicina, Centro Universitário de Belo Horizonte, Belo Horizonte, Brazil
| | | |
Collapse
|
8
|
Song R, Lei S, Yang S, Wu SJ. LncRNA PAXIP1-AS1 fosters the pathogenesis of pulmonary arterial hypertension via ETS1/WIPF1/RhoA axis. J Cell Mol Med 2021; 25:7321-7334. [PMID: 34245091 PMCID: PMC8335679 DOI: 10.1111/jcmm.16761] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 05/14/2021] [Accepted: 05/24/2021] [Indexed: 12/17/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a life‐threatening disease featured with elevated pulmonary vascular resistance and progressive pulmonary vascular remodelling. It has been demonstrated that lncRNA PAXIP1‐AS1 could influence the transcriptome in PAH. However, the exact molecular mechanism of PAXIP1‐AS1 in PAH pathogenesis remains largely unknown. In this study, in vivo rat PAH model was established by monocrotaline (MCT) induction and hypoxia was used to induce in vitro PAH model using human pulmonary artery smooth muscle cells (hPASMCs). Histological examinations including H&E, Masson's trichrome staining and immunohistochemistry were subjected to evaluate the pathological changes of lung tissues. Expression patterns of PAXIP1‐AS1 and RhoA were assessed using qRT‐PCR and Western blotting, respectively. CCK‐8, BrdU assay and immunofluorescence of Ki67 were performed to measure the cell proliferation. Wound healing and transwell assays were employed to evaluate the capacity of cell migration. Dual‐luciferase reporter assay, co‐immunoprecipitation, RIP and CHIP assays were employed to verify the PAXIP1‐AS1/ETS1/WIPF1/RhoA regulatory network. It was found that the expression of PAXIP1‐AS1 and RhoA was remarkably higher in both lung tissues and serum of MCT‐induced PAH rats, as well as in hypoxia‐induced hPASMCs. PAXIP1‐AS1 knockdown remarkably suppressed hypoxia‐induced cell viability and migration of hPASMCs. PAXIP1‐AS1 positively regulated WIPF1 via recruiting transcriptional factor ETS1, of which knockdown reversed PAXIP1‐AS1‐mediated biological functions. Co‐immunoprecipitation validated the WIPF1/RhoA interaction. In vivo experiments further revealed the role of PAXIP1‐AS1 in PAH pathogenesis. In summary, lncRNA PAXIP1‐AS1 promoted cell viability and migration of hPASMCs via ETS1/WIPF1/RhoA, which might provide a potential therapeutic target for PAH treatment.
Collapse
Affiliation(s)
- Rong Song
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Si Lei
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Song Yang
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shang-Jie Wu
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Mprah R, Adzika GK, Gyasi YI, Ndzie Noah ML, Adu-Amankwaah J, Adekunle AO, Duah M, Wowui PI, Weili Q. Glutaminolysis: A Driver of Vascular and Cardiac Remodeling in Pulmonary Arterial Hypertension. Front Cardiovasc Med 2021; 8:667446. [PMID: 33996951 PMCID: PMC8113389 DOI: 10.3389/fcvm.2021.667446] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 03/08/2021] [Indexed: 12/30/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a decimating ailment described by chronic precapillary pulmonary hypertension, an elevated mean pulmonary arterial pressure with a normal pulmonary capillary wedge pressure, and a raised pulmonary vascular resistance resulting in increased right ventricular afterload culminating in heart failure and death. Current PAH treatments regulate the vasodilatory/vasoconstrictory balance of pulmonary vessels. However, these treatment options are unable to stop the progression of, or reverse, an already established disease. Recent studies have advanced a metabolic dysregulation, featuring increased glutamine metabolism, as a mechanism driving PAH progression. Metabolic dysregulation in PAH leads to increased glutaminolysis to produce substrate to meet the high-energy requirement by hyperproliferative and apoptosis-resistant pulmonary vascular cells. This article explores the role of glutamate metabolism in PAH and how it could be targeted as an anti-remodeling therapeutic strategy.
Collapse
Affiliation(s)
- Richard Mprah
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | | | - Yusif I. Gyasi
- Department of Chemistry & Biochemistry, Central Michigan University, Mount Pleasant, TX, United States
| | | | | | | | - Maxwell Duah
- Haematology Department, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | | | - Qiao Weili
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
10
|
Pulmonary Hypertension in Acute and Chronic High Altitude Maladaptation Disorders. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18041692. [PMID: 33578749 PMCID: PMC7916528 DOI: 10.3390/ijerph18041692] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 12/13/2022]
Abstract
Alveolar hypoxia is the most prominent feature of high altitude environment with well-known consequences for the cardio-pulmonary system, including development of pulmonary hypertension. Pulmonary hypertension due to an exaggerated hypoxic pulmonary vasoconstriction contributes to high altitude pulmonary edema (HAPE), a life-threatening disorder, occurring at high altitudes in non-acclimatized healthy individuals. Despite a strong physiologic rationale for using vasodilators for prevention and treatment of HAPE, no systematic studies of their efficacy have been conducted to date. Calcium-channel blockers are currently recommended for drug prophylaxis in high-risk individuals with a clear history of recurrent HAPE based on the extensive clinical experience with nifedipine in HAPE prevention in susceptible individuals. Chronic exposure to hypoxia induces pulmonary vascular remodeling and development of pulmonary hypertension, which places an increased pressure load on the right ventricle leading to right heart failure. Further, pulmonary hypertension along with excessive erythrocytosis may complicate chronic mountain sickness, another high altitude maladaptation disorder. Importantly, other causes than hypoxia may potentially underlie and/or contribute to pulmonary hypertension at high altitude, such as chronic heart and lung diseases, thrombotic or embolic diseases. Extensive clinical experience with drugs in patients with pulmonary arterial hypertension suggests their potential for treatment of high altitude pulmonary hypertension. Small studies have demonstrated their efficacy in reducing pulmonary artery pressure in high altitude residents. However, no drugs have been approved to date for the therapy of chronic high altitude pulmonary hypertension. This work provides a literature review on the role of pulmonary hypertension in the pathogenesis of acute and chronic high altitude maladaptation disorders and summarizes current knowledge regarding potential treatment options.
Collapse
|
11
|
Xin J, Zhang H, He Y, Duren Z, Bai C, Chen L, Luo X, Yan DS, Zhang C, Zhu X, Yuan Q, Feng Z, Cui C, Qi X, Ouzhuluobu, Wong WH, Wang Y, Su B. Chromatin accessibility landscape and regulatory network of high-altitude hypoxia adaptation. Nat Commun 2020; 11:4928. [PMID: 33004791 PMCID: PMC7529806 DOI: 10.1038/s41467-020-18638-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/03/2020] [Indexed: 12/27/2022] Open
Abstract
High-altitude adaptation of Tibetans represents a remarkable case of natural selection during recent human evolution. Previous genome-wide scans found many non-coding variants under selection, suggesting a pressing need to understand the functional role of non-coding regulatory elements (REs). Here, we generate time courses of paired ATAC-seq and RNA-seq data on cultured HUVECs under hypoxic and normoxic conditions. We further develop a variant interpretation methodology (vPECA) to identify active selected REs (ASREs) and associated regulatory network. We discover three causal SNPs of EPAS1, the key adaptive gene for Tibetans. These SNPs decrease the accessibility of ASREs with weakened binding strength of relevant TFs, and cooperatively down-regulate EPAS1 expression. We further construct the downstream network of EPAS1, elucidating its roles in hypoxic response and angiogenesis. Collectively, we provide a systematic approach to interpret phenotype-associated noncoding variants in proper cell types and relevant dynamic conditions, to model their impact on gene regulation.
Collapse
Affiliation(s)
- Jingxue Xin
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
- Bio-X Program, Stanford University, Stanford, CA, 94305, USA
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Hui Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
| | - Yaoxi He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Zhana Duren
- Departments of Statistics, Stanford University, Stanford, CA, 94305, USA
- Center for Human Genetics and Department of Genetics and Biochemistry, Clemson University, Greenwood, SC, 29646, USA
| | - Caijuan Bai
- High Altitude Medical Research Center, School of Medicine, Tibetan University, 850000, Lhasa, China
| | - Lang Chen
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Xin Luo
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Dong-Sheng Yan
- School of Mathematical Science, Inner Mongolia University, 010021, Huhhot, China
| | - Chaoyu Zhang
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Xiang Zhu
- Departments of Statistics, Stanford University, Stanford, CA, 94305, USA
| | - Qiuyue Yuan
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Zhanying Feng
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Chaoying Cui
- High Altitude Medical Research Center, School of Medicine, Tibetan University, 850000, Lhasa, China
| | - Xuebin Qi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China
| | - Ouzhuluobu
- High Altitude Medical Research Center, School of Medicine, Tibetan University, 850000, Lhasa, China
| | - Wing Hung Wong
- Bio-X Program, Stanford University, Stanford, CA, 94305, USA.
- Departments of Statistics, Stanford University, Stanford, CA, 94305, USA.
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Yong Wang
- CEMS, NCMIS, MDIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, 100190, Beijing, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China.
- University of Chinese Academy of Sciences, 100101, Beijing, China.
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 330106, Hangzhou, China.
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China.
| |
Collapse
|
12
|
Nitulescu GM, Paunescu H, Moschos SA, Petrakis D, Nitulescu G, Ion GND, Spandidos DA, Nikolouzakis TK, Drakoulis N, Tsatsakis A. Comprehensive analysis of drugs to treat SARS‑CoV‑2 infection: Mechanistic insights into current COVID‑19 therapies (Review). Int J Mol Med 2020; 46:467-488. [PMID: 32468014 PMCID: PMC7307820 DOI: 10.3892/ijmm.2020.4608] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 05/18/2020] [Indexed: 12/16/2022] Open
Abstract
The major impact produced by the severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) focused many researchers attention to find treatments that can suppress transmission or ameliorate the disease. Despite the very fast and large flow of scientific data on possible treatment solutions, none have yet demonstrated unequivocal clinical utility against coronavirus disease 2019 (COVID‑19). This work represents an exhaustive and critical review of all available data on potential treatments for COVID‑19, highlighting their mechanistic characteristics and the strategy development rationale. Drug repurposing, also known as drug repositioning, and target based methods are the most used strategies to advance therapeutic solutions into clinical practice. Current in silico, in vitro and in vivo evidence regarding proposed treatments are summarized providing strong support for future research efforts.
Collapse
Affiliation(s)
| | - Horia Paunescu
- Faculty of Medicine, ′Carol Davila′ University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Sterghios A. Moschos
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University
- PulmoBioMed Ltd., Newcastle-Upon-Tyne NE1 8ST, UK
| | | | | | | | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion
| | | | - Nikolaos Drakoulis
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | | |
Collapse
|
13
|
Sandoval J, Del Valle-Mondragón L, Masso F, Zayas N, Pulido T, Teijeiro R, Gonzalez-Pacheco H, Olmedo-Ocampo R, Sisniega C, Paez-Arenas A, Pastelin-Hernandez G, Gomez-Arroyo J, Voelkel NF. Angiotensin converting enzyme 2 and angiotensin (1-7) axis in pulmonary arterial hypertension. Eur Respir J 2020; 56:13993003.02416-2019. [PMID: 32241831 DOI: 10.1183/13993003.02416-2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/21/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND In animal models of pulmonary arterial hypertension (PAH), angiotensin-converting enzyme (ACE)2 and angiotensin (Ang)-(1-7) have been shown to have vasodilatory, antiproliferative, antifibrotic and antihypertrophic properties. However, the status and role of the ACE2-Ang(1-7) axis in human PAH is incompletely understood. METHODS We studied 85 patients with a diagnosis of PAH of distinct aetiologies. 55 healthy blood donors paired for age and sex served as controls. Blood samples were obtained from the pulmonary artery in patients with PAH during right heart catheterisation. Peripheral blood was obtained for both groups. Ang(1-7) and -II were measured using zone capillary electrophoresis. Aldosterone, Ang(1-9), AngA and ACE2 were measured using ELISA, and ACE2 activity was determined enzymatically. RESULTS Of the 85 patients, 47 had idiopathic PAH, 25 had PAH associated with congenital heart disease and 13 had PAH associated with collagen vascular disease. Compared to controls, patients with PAH had a higher concentration of AngII (median 1.03, interquartile range 0.72-1.88 pmol·mL-1 versus 0.19, 0.10-0.37 pmol·mL-1; p<0.001) and of aldosterone (88.7, 58.7-132 ng·dL-1 versus 12.9, 9.55-19.9 ng·dL-1; p<0.001). Conversely, PAH patients had a lower concentration of Ang(1-7) than controls (0.69, 0.474-0.91 pmol·mL-1 versus 4.07, 2.82-6.73 pmol·mL-1; p<0.001), and a lower concentration of Ang(1-9) and AngA. Similarly, the ACE2 concentration was higher than in controls (8.7, 5.35-13.2 ng·mL-1 versus 4.53, 1.47-14.3 ng·mL-1; p=0.011), whereas the ACE2 activity was significantly reduced (1.88, 1.08-2.81 nmol·mL-1 versus 5.97, 3.1-17.8 nmol·mL-1; p<0.001). No significant differences were found among the three different aetiological forms of PAH. CONCLUSIONS The AngII-ACE2-Ang(1-7) axis appears to be altered in human PAH and we propose that this imbalance, in favour of AngII, plays a role in the pathogenesis of the severe PAH. Further mechanistic studies are warranted.
Collapse
Affiliation(s)
- Julio Sandoval
- Cardiopulmonary Dept, Instituto Nacional de Cardiologia, Mexico City, Mexico
| | | | - Felipe Masso
- Physiology and Molecular Biology Dept of the "Ignacio Chávez", National Institute of Cardiology, Mexico City, Mexico
| | - Nayeli Zayas
- Cardiopulmonary Dept, Instituto Nacional de Cardiologia, Mexico City, Mexico
| | - Tomás Pulido
- Cardiopulmonary Dept, Instituto Nacional de Cardiologia, Mexico City, Mexico
| | - Ricardo Teijeiro
- Cardiopulmonary Dept, Instituto Nacional de Cardiologia, Mexico City, Mexico
| | | | | | - Carlos Sisniega
- Cardiopulmonary Dept, Instituto Nacional de Cardiologia, Mexico City, Mexico
| | - Araceli Paez-Arenas
- Physiology and Molecular Biology Dept of the "Ignacio Chávez", National Institute of Cardiology, Mexico City, Mexico
| | | | - Jose Gomez-Arroyo
- Cardiopulmonary Dept, Instituto Nacional de Cardiologia, Mexico City, Mexico.,Division of Pulmonary and Critical Care Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Norbert F Voelkel
- Dept of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Feczko AF, Raja S. Commentary: The pressure to treat pulmonary artery hypertension. J Thorac Cardiovasc Surg 2020; 161:e435-e436. [PMID: 32690412 DOI: 10.1016/j.jtcvs.2020.04.131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 10/24/2022]
Affiliation(s)
- Andrew F Feczko
- Department of Thoracic and Cardiovascular Surgery, Heart, Vascular, and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Siva Raja
- Department of Thoracic and Cardiovascular Surgery, Heart, Vascular, and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, Ohio.
| |
Collapse
|
15
|
Spaczyńska M, Rocha SF, Oliver E. Pharmacology of Pulmonary Arterial Hypertension: An Overview of Current and Emerging Therapies. ACS Pharmacol Transl Sci 2020; 3:598-612. [PMID: 32832865 DOI: 10.1021/acsptsci.0c00048] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Indexed: 12/21/2022]
Abstract
Pulmonary arterial hypertension is a rare and devastating disease characterized by an abnormal chronic increase in pulmonary arterial pressure above 20 mmHg at rest, with a poor prognosis if not treated. Currently, there is not a single fully effective therapy, even though a dozen of drugs have been developed in the last decades. Pulmonary arterial hypertension is a multifactorial disease, meaning that several molecular mechanisms are implicated in its pathology. The main molecular pathways regulating the pulmonary vasomotor tone-endothelin, nitric oxide, and prostacyclin-are the most biologically and therapeutically explored to date. However, drugs targeting these pathways have already found their limitations. In the last years, translational research and clinical trials have made a strong effort in suggesting and testing novel therapeutic strategies for this disease. These approaches involve targeting the main molecular pathways with novel drugs, drug repurposing for novel targets, and also using combinatorial therapies. In this review, we summarize current strategies and drugs targeting the endothelin, nitric oxide, and prostacyclin pathways, as well as, the emerging new drugs proposed to cope with vascular remodelling, metabolic switch, perivascular inflammation, epigenetic modifications, estrogen deregulation, serotonin, and other neurohumoral mechanisms characteristic of this disease. Nowadays, pulmonary arterial hypertension remains an incurable disease; however, the incoming new knowledge makes us believe that new promising therapies are coming to the clinical arena soon.
Collapse
Affiliation(s)
- Monika Spaczyńska
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Susana F Rocha
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Eduardo Oliver
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, 28029, Spain.,Centro de Investigaciones Biomédicas en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, 28029, Spain
| |
Collapse
|
16
|
Kay D, Kadri F, Fitzpatrick G, Alnuaimat H, Reddy R, Ataya A. Anti-synthetase syndrome-associated pulmonary veno-occlusive disease. Pulm Circ 2020; 10:2045894020935289. [PMID: 32655855 PMCID: PMC7331768 DOI: 10.1177/2045894020935289] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 05/28/2020] [Indexed: 12/28/2022] Open
Abstract
Pulmonary arterial hypertension has been reported with a prevalence of 7.9% in patients with anti-synthetase syndrome; however, anti-synthetase syndrome associated with pulmonary veno-occlusive disease (PVOD) has never before been described in the literature. We present a novel case of anti-synthetase syndrome-associated PVOD in a patient who presented with hypoxic respiratory failure associated with right heart failure and was diagnosed with anti-synthetase syndrome based on his autoimmune serology and pre-capillary pulmonary hypertension on right heart catheterization. He was initiated on pulmonary arterial hypertension therapy, but with escalating dose of parenteral epoprostenol, experienced acute clinical worsening with chest imaging concerning for PVOD that was confirmed on autopsy. Anti-synthetase syndrome can be associated with PVOD, and it should be suspected in patients who have evidence of pre-capillary pulmonary hypertension and who deteriorate with the initiation of pulmonary hypertension-specific therapy.
Collapse
Affiliation(s)
- Diana Kay
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Ferdous Kadri
- Department of Internal Medicine, University of Florida, Gainesville, FL, USA
| | | | - Hassan Alnuaimat
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Raju Reddy
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Ali Ataya
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
17
|
Gomez-Puerto MC, Sun XQ, Schalij I, Orriols M, Pan X, Szulcek R, Goumans MJ, Bogaard HJ, Zhou Q, ten Dijke P. MnTBAP Reverses Pulmonary Vascular Remodeling and Improves Cardiac Function in Experimentally Induced Pulmonary Arterial Hypertension. Int J Mol Sci 2020; 21:E4130. [PMID: 32531895 PMCID: PMC7312610 DOI: 10.3390/ijms21114130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 12/18/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by obstructed pulmonary vasculatures. Current therapies for PAH are limited and only alleviate symptoms. Reduced levels of BMPR2 are associated with PAH pathophysiology. Moreover, reactive oxygen species, inflammation and autophagy have been shown to be hallmarks in PAH. We previously demonstrated that MnTBAP, a synthetic metalloporphyrin with antioxidant and anti-inflammatory activity, inhibits the turn-over of BMPR2 in human umbilical vein endothelial cells. Therefore, we hypothesized that MnTBAP might be used to treat PAH. Human pulmonary artery endothelial cells (PAECs), as well as pulmonary microvascular endothelial (MVECs) and smooth muscle cells (MVSMCs) from PAH patients, were treated with MnTBAP. In vivo, either saline or MnTBAP was given to PAH rats induced by Sugen 5416 and hypoxia (SuHx). On PAECs, MnTBAP was found to increase BMPR2 protein levels by blocking autophagy. Moreover, MnTBAP increased BMPR2 levels in pulmonary MVECs and MVSMCs isolated from PAH patients. In SuHx rats, MnTBAP reduced right ventricular (RV) afterload by reversing pulmonary vascular remodeling, including both intima and media layers. Furthermore, MnTBAP improved RV function and reversed RV dilation in SuHx rats. Taken together, these data highlight the importance of MnTBAP as a potential therapeutic treatment for PAH.
Collapse
Affiliation(s)
- Maria Catalina Gomez-Puerto
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (M.C.G.-P.); (M.O.); (M.-J.G.); (P.t.D.)
| | - Xiao-Qing Sun
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (X.-Q.S.); (I.S.); (X.P.); (R.S.)
| | - Ingrid Schalij
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (X.-Q.S.); (I.S.); (X.P.); (R.S.)
| | - Mar Orriols
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (M.C.G.-P.); (M.O.); (M.-J.G.); (P.t.D.)
| | - Xiaoke Pan
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (X.-Q.S.); (I.S.); (X.P.); (R.S.)
| | - Robert Szulcek
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (X.-Q.S.); (I.S.); (X.P.); (R.S.)
| | - Marie-José Goumans
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (M.C.G.-P.); (M.O.); (M.-J.G.); (P.t.D.)
| | - Harm-Jan Bogaard
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (X.-Q.S.); (I.S.); (X.P.); (R.S.)
| | - Qian Zhou
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
| | - Peter ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (M.C.G.-P.); (M.O.); (M.-J.G.); (P.t.D.)
| |
Collapse
|
18
|
Abstract
Investigations into the mixed muscle-secretory phenotype of cardiomyocytes from the atrial appendages of the heart led to the discovery that these cells produce, in a regulated manner, two polypeptide hormones - the natriuretic peptides - referred to as atrial natriuretic factor or atrial natriuretic peptide (ANP) and brain or B-type natriuretic peptide (BNP), thereby demonstrating an endocrine function for the heart. Studies on the gene encoding ANP (NPPA) initiated the field of modern research into gene regulation in the cardiovascular system. Additionally, ANP and BNP were found to be the natural ligands for cell membrane-bound guanylyl cyclase receptors that mediate the effects of natriuretic peptides through the generation of intracellular cGMP, which interacts with specific enzymes and ion channels. Natriuretic peptides have many physiological actions and participate in numerous pathophysiological processes. Important clinical entities associated with natriuretic peptide research include heart failure, obesity and systemic hypertension. Plasma levels of natriuretic peptides have proven to be powerful diagnostic and prognostic biomarkers of heart disease. Development of pharmacological agents that are based on natriuretic peptides is an area of active research, with vast potential benefits for the treatment of cardiovascular disease.
Collapse
|
19
|
Miao H, Qiu F, Zhu L, Jiang B, Yuan Y, Huang B, Zhang Y. Novel angiogenesis strategy to ameliorate pulmonary hypertension. J Thorac Cardiovasc Surg 2020; 161:e417-e434. [PMID: 32359908 DOI: 10.1016/j.jtcvs.2020.03.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/28/2020] [Accepted: 03/14/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To select a suitable combination of classic angiogenic and vascular stabilization factors to improve the proliferation and maturity of neovascularization of lung tissue in a rat pulmonary arterial hypertension (PAH) model. METHODS PAH rat model was established by intraperitoneal injection of monocrotaline. Proangiogenic factors hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF), as well as vascular stabilization factors angiopoietin-1 (Ang-1), platelet-derived growth factor, and transforming growth factor-beta were transfected by pairs into the lung tissue of rats with PAH through lentivirus. Four weeks later, pulmonary artery angiography and hemodynamic parameters were determined to testify the remission of PAH. Immunofluorescence staining and Western blot were performed to investigate the structure and function of neovascularization. RESULTS The pulmonary artery pressure and weight index of the right ventricle in HGF+Ang-1 and VEGF+Ang-1 groups were significantly decreased compared with vehicle group. The contrast medium filling time and right pulmonary artery root diameter were also significantly decreased. In addition, the maturity and perfusion of neovascularization in HGF+Ang-1 and VEGF+Ang-1 groups were promoted compared to vehicle group, and vascular leakage was reduced. Finally, the adherens junction integrity of vascular endothelial cells in HGF+Ang-1 and VEGF+Ang-1 combinations was upregulated compared with other combinations. CONCLUSIONS HGF+Ang-1 transfection and VEGF+Ang-1 transfection alleviate PAH by promoting maturation and stability of new blood vessels, which may be potential candidates for PAH treatment.
Collapse
Affiliation(s)
- Haoran Miao
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China; Department of Thoracic Cardiovascular Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fan Qiu
- Department of Thoracic Cardiovascular Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Thoracic Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lidong Zhu
- Department of Thoracic Cardiovascular Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bo Jiang
- Department of Thoracic Cardiovascular Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanliang Yuan
- Department of Thoracic Cardiovascular Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bing Huang
- Department of Thoracic Cardiovascular Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Thoracic Cardiovascular Surgery, Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yiqian Zhang
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China.
| |
Collapse
|
20
|
YAO L, YANG YX, CAO H, REN HH, NIU Z, SHI L. Osthole attenuates pulmonary arterial hypertension by the regulation of sphingosine 1-phosphate in rats. Chin J Nat Med 2020; 18:308-320. [DOI: 10.1016/s1875-5364(20)30038-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Indexed: 10/24/2022]
|
21
|
Yan G, Wang J, Yi T, Cheng J, Guo H, He Y, Shui X, Wu Z, Huang S, Lei W. Baicalin prevents pulmonary arterial remodeling in vivo via the AKT/ERK/NF-κB signaling pathways. Pulm Circ 2019; 9:2045894019878599. [PMID: 31723406 PMCID: PMC6831981 DOI: 10.1177/2045894019878599] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 09/05/2019] [Indexed: 02/06/2023] Open
Abstract
Pulmonary arterial hypertension is a rapidly progressive and often fatal disease. As the pathogenesis of pulmonary arterial hypertension remains unclear, there is currently no good drug for pulmonary arterial hypertension and new therapy is desperately needed. This study investigated the effects and mechanism of baicalin on vascular remodeling in rats with pulmonary arterial hypertension. A rat pulmonary arterial hypertension model was constructed using intraperitoneal injection of monocrotaline, and different doses of baicalin were used to treat these rats. The mean pulmonary arterial pressure (mPAP) and right ventricular systolic pressure (RVSP) were measured with a right heart catheter. Moreover, the hearts were dissected to determine the right ventricular hypertrophy index (RVHI). The lung tissues were stained with H&E and Masson's staining to estimate the pulmonary vascular remodeling and collagen fibrosis, and the expression of proteins in the AKT, ERK, and NF-κB p65 phosphorylation (p-AKT, p-ERK, p-p65) was examined by Western blot analysis. We found that compared with untreated pulmonary arterial hypertension rats, baicalin ameliorated pulmonary vascular remodeling and cardiorespiratory injury, inhibited p-p65 and p-ERK expression, and promoted p-AKT and p-eNOS expression. In conclusion, baicalin interfered with pulmonary vascular remodeling and pulmonary arterial hypertension development in rats through the AKT/eNOS, ERK and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Guosen Yan
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, China.,Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jinxia Wang
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, China.,Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Tao Yi
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, China.,Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Junfen Cheng
- Department of Respiration, the Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Haixu Guo
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, China.,Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yuan He
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, China
| | - Xiaorong Shui
- Laboratory of Vascular Surgery, Guangdong Medical University, Zhanjiang, China
| | - Zeyong Wu
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shian Huang
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Wei Lei
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, China.,Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
22
|
Gomez‐Puerto MC, van Zuijen I, Huang CJZ, Szulcek R, Pan X, van Dinther MAH, Kurakula K, Wiesmeijer CC, Goumans M, Bogaard H, Morrell NW, Rana AA, ten Dijke P. Autophagy contributes to BMP type 2 receptor degradation and development of pulmonary arterial hypertension. J Pathol 2019; 249:356-367. [PMID: 31257577 PMCID: PMC6852495 DOI: 10.1002/path.5322] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 06/05/2019] [Accepted: 06/24/2019] [Indexed: 12/21/2022]
Abstract
Pulmonary arterial hypertension (PAH) is characterised by an increase in mean pulmonary arterial pressure which almost invariably leads to right heart failure and premature death. More than 70% of familial PAH and 20% of idiopathic PAH patients carry heterozygous mutations in the bone morphogenetic protein (BMP) type 2 receptor (BMPR2). However, the incomplete penetrance of BMPR2 mutations suggests that other genetic and environmental factors contribute to the disease. In the current study, we investigate the contribution of autophagy in the degradation of BMPR2 in pulmonary vascular cells. We demonstrate that endogenous BMPR2 is degraded through the lysosome in primary human pulmonary artery endothelial (PAECs) and smooth muscle cells (PASMCs): two cell types that play a key role in the pathology of the disease. By means of an elegant HaloTag system, we show that a block in lysosomal degradation leads to increased levels of BMPR2 at the plasma membrane. In addition, pharmacological or genetic manipulations of autophagy allow us to conclude that autophagy activation contributes to BMPR2 degradation. It has to be further investigated whether the role of autophagy in the degradation of BMPR2 is direct or through the modulation of the endocytic pathway. Interestingly, using an iPSC-derived endothelial cell model, our findings indicate that BMPR2 heterozygosity alone is sufficient to cause an increased autophagic flux. Besides BMPR2 heterozygosity, pro-inflammatory cytokines also contribute to an augmented autophagy in lung vascular cells. Furthermore, we demonstrate an increase in microtubule-associated protein 1 light chain 3 beta (MAP1LC3B) levels in lung sections from PAH induced in rats. Accordingly, pulmonary microvascular endothelial cells (MVECs) from end-stage idiopathic PAH patients present an elevated autophagic flux. Our findings support a model in which an increased autophagic flux in PAH patients contributes to a greater decrease in BMPR2 levels. Altogether, this study sheds light on the basic mechanisms of BMPR2 degradation and highlights a crucial role for autophagy in PAH. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Arterial Pressure
- Autophagy
- Bone Morphogenetic Protein Receptors, Type II/genetics
- Bone Morphogenetic Protein Receptors, Type II/metabolism
- Cell Line
- Cytokines/metabolism
- Disease Models, Animal
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Female
- Heterozygote
- Humans
- Inflammation Mediators/metabolism
- Lysosomes/metabolism
- Lysosomes/pathology
- Male
- Microtubule-Associated Proteins/metabolism
- Middle Aged
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Proteolysis
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/pathology
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Rats
- Signal Transduction
- Young Adult
Collapse
Affiliation(s)
- Maria Catalina Gomez‐Puerto
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | - Iris van Zuijen
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | | | - Robert Szulcek
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pulmonary MedicineAmsterdam Cardiovascular SciencesAmsterdamThe Netherlands
| | - Xiaoke Pan
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pulmonary MedicineAmsterdam Cardiovascular SciencesAmsterdamThe Netherlands
| | - Maarten AH van Dinther
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | - Kondababu Kurakula
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | - Catharina C Wiesmeijer
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | - Marie‐Jose Goumans
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | - Harm‐Jan Bogaard
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pulmonary MedicineAmsterdam Cardiovascular SciencesAmsterdamThe Netherlands
| | | | | | - Peter ten Dijke
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
23
|
Wang Z, Jiang X, Zhang X, Tian G, Yang R, Wu J, Zou X, Liu Z, Yang X, Wu C, Shi J, Li J, Suo J, Wang Y, Zhang R, Xu Z, Gong X, He Y, Zhu W, Aisa HA, Jiang H, Xu Y, Shen J. Pharmacokinetics-Driven Optimization of 4(3 H)-Pyrimidinones as Phosphodiesterase Type 5 Inhibitors Leading to TPN171, a Clinical Candidate for the Treatment of Pulmonary Arterial Hypertension. J Med Chem 2019; 62:4979-4990. [PMID: 31021628 DOI: 10.1021/acs.jmedchem.9b00123] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Phosphodiesterase type 5 (PDE5) inhibitors are first-line therapy for pulmonary arterial hypertension (PAH) and erectile dysfunction. As a continuing work to improve the terminal half-lives and oral bioavailabilities of our previously reported 4(3 H)-pyrimidones, a pharmacokinetics-driven optimization focusing on the terminal substituent is described. Two major congeneric series of 4(3 H)-pyrimidones, the aminosulfonylphenylpyrimidones and acylaminophenylpyrimidones, were designed, synthesized, and pharmacologically assessed in vitro and in vivo. Among them, compound 15 (TPN171) with subnanomolar potency for PDE5 and good selectivity over PDE6 was finally recognized as a potential drug candidate, and its pharmacokinetic profiles in rats and dogs are significantly improved compared to the starting compound (3). Moreover, TPN171 was proven to exert a longer lasting effect than sildenafil in animal models, providing a foundation for a once-daily oral administration for its clinical use. TPN171 is currently being investigated in a phase II clinical trial for the treatment of PAH.
Collapse
Affiliation(s)
- Zhen Wang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Xiangrui Jiang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Xianglei Zhang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China.,School of Pharmacy , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Guanghui Tian
- Vigonvita Life Science Co., Ltd. , Suzhou 215123 , China
| | - Rulei Yang
- Vigonvita Life Science Co., Ltd. , Suzhou 215123 , China
| | - Jianzhong Wu
- Vigonvita Life Science Co., Ltd. , Suzhou 215123 , China
| | - Xiaoli Zou
- Vigonvita Life Science Co., Ltd. , Suzhou 215123 , China
| | - Zheng Liu
- Topharman Shanghai Co., Ltd. , Shanghai 201203 , China
| | - Xiaojun Yang
- Topharman Shanghai Co., Ltd. , Shanghai 201203 , China
| | - Chunhui Wu
- Topharman Shanghai Co., Ltd. , Shanghai 201203 , China
| | - Jing Shi
- Topharman Shanghai Co., Ltd. , Shanghai 201203 , China
| | - Jianfeng Li
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Jin Suo
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Yu Wang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Rongxia Zhang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Zhijian Xu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Xudong Gong
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China.,Key Laboratory of Plant Resources and Chemistry in Arid Regions , Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Urumqi 830011 , China
| | - Yang He
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China
| | - Weiliang Zhu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China.,School of Pharmacy , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Haji Akber Aisa
- Key Laboratory of Plant Resources and Chemistry in Arid Regions , Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences , Urumqi 830011 , China
| | - Hualiang Jiang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China.,School of Pharmacy , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Yechun Xu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China.,School of Pharmacy , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Jingshan Shen
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai 201203 , China.,School of Pharmacy , University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
24
|
Wang L, Wang Y, Lei Z. Chrysin ameliorates ANTU-induced pulmonary edema and pulmonary arterial hypertension via modulation of VEGF and eNOs. J Biochem Mol Toxicol 2019; 33:e22332. [PMID: 30974023 DOI: 10.1002/jbt.22332] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/19/2019] [Accepted: 03/15/2019] [Indexed: 12/26/2022]
Abstract
Alpha-naphthylthiourea (ANTU), a rodenticide induces lung toxicity. Chrysin a flavonoid possesses antioxidant, anti-inflammatory, and antihypertensive potential. The aim of this study was to evaluate the efficacy of chrysin against ANTU-induced pulmonary edema (PE) and pulmonary arterial hypertension (PAH) in laboratory rats. Sprague-Dawley rats were used to induce PE (ANTU, 10 mg/kg, ip) and PAH (ANTU, 5 mg/kg, ip, 4 weeks). Animals were treated with chrysin (10, 20, and 40 mg/kg) and various biochemical, molecular, and histological parameters were evaluated. Acute administration of ANTU induces PE revealed by significant (P < 0.05) increase in relative lung weight, pleural effusion volume, lung edema, bronchoalveolar lavage fluid cell counts, total protein, 5-hydroxytryptamine (5-HT), lactate dehydrogenase (LDH), and γ-glutamyl transferase (GGT), whereas pretreatment with chrysin (20 and 40 mg/kg, ip) significantly (P < 0.05) attenuated these ANTU-induced biochemical and histological alterations. Repeated administration of ANTU caused induction of PAH evaluated by significant (P < 0.05) alterations in electrocardiographic, hemodynamic changes, and left ventricular function, whereas chrysin (20 and 40 mg/kg, p.o.) treatment significantly (P < 0.05) attenuated these alterations. ANTU-induced hematological and serum biochemical (aspartate transaminase, alanine transaminase, LDH, and creatinine kinase MB) alterations were significantly (P < 0.05) inhibited by chrysin. It also significantly (P < 0.05) decreased elevated levels of oxido-nitrosative stress in the right ventricle (RV) and lung. Chrysin significantly (P < 0.05) attenuated downregulated endothelial nitric oxide synthase and upregulated vascular endothelial growth factor messenger RNA and protein expressions both in the RV and pulmonary artery. Chrysin inhibited ANTU-induced PE and PAH via modulation of inflammatory responses (5-HT, LDH, and GGT), oxido-nitrosative stress, and VEGF and eNOs levels.
Collapse
Affiliation(s)
- Linlin Wang
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Ye Wang
- Department of Paediatrics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Zhang Lei
- Department of Neurology, the Second Clinical Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|