1
|
Lu G, Du R, Lu L, Wang Q, Zhang M, Gu X, Feng N, Zhang S, Liu Y, Li J, Pei J. Macrophage-specific κ-OR knockout exacerbates inflammation in hypoxic pulmonary hypertension. Eur J Pharmacol 2025; 986:177152. [PMID: 39586395 DOI: 10.1016/j.ejphar.2024.177152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Hypoxic pulmonary hypertension (HPH), a prevalent subtype of pulmonary arterial hypertension, is characterized by pulmonary vasoconstriction (HPV) and vascular remodeling, accompanied by inflammatory responses. Recent in vivo studies have shown a critical role of the κ-opioid receptor (κ-OR) in modulating the aforementioned pathological processes. Specifically, macrophage-specific κ-OR-knockout models have shown inflammatory response exacerbation with pulmonary hypertension and vascular remodeling. Conversely, the novel κ-OR agonist Q-U50, 488H inhibits inflammatory pathways, thereby attenuating pulmonary vasoconstriction and vascular remodeling. The present study revealed that hypoxia promoted macrophage infiltration and pulmonary artery smooth muscle cell proliferation. Moreover, under these conditions, macrophages secreted interleukin (IL)-6, which triggered the signal transducer and activator of transcription 3 (STAT3)/miR-153-3p signaling cascade. Herein, we identified miR-153-3p downregulated κ-OR gene expression, which is a key contributor to HPV and remodeling, it was identified as a pivotal regulator of κ-OR mRNA levels. The pharmacological activation of κ-OR inhibited IL-6 release from macrophages and disrupted the IL-6/STAT3/miR-153-3p pathway. This dual action of κ-OR activation mitigated pulmonary artery contraction and remodeling, thereby offering a protective mechanism against HPH. The present findings have delineated a novel negative feedback loop driving HPH pathogenesis and suggested that targeting the κ-OR-IL-6-STAT3-miR-153-3p axis represented a promising therapeutic strategy against HPH.
Collapse
MESH Headings
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/agonists
- Animals
- Macrophages/metabolism
- MicroRNAs/genetics
- STAT3 Transcription Factor/metabolism
- STAT3 Transcription Factor/genetics
- Interleukin-6/metabolism
- Interleukin-6/genetics
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/pathology
- Hypoxia/complications
- Hypoxia/genetics
- Mice
- Mice, Knockout
- Inflammation/genetics
- Inflammation/pathology
- Pulmonary Artery/pathology
- Signal Transduction
- Male
- Mice, Inbred C57BL
- Vascular Remodeling/drug effects
- Vascular Remodeling/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Cell Proliferation/drug effects
- Vasoconstriction/drug effects
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology
Collapse
Affiliation(s)
- Guofang Lu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China; State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Rui Du
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Linhe Lu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Qiaojuan Wang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Min Zhang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaoming Gu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Na Feng
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Shumiao Zhang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yali Liu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Juan Li
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jianming Pei
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
2
|
Bajwa EK, Cislak D, Kumar A, Li D, Messina EJ, Reynders T, Denef JF, Corcea V, Buch KP, Lai E, Stoch SA. Phase 1 Study of MK-5475, an Inhaled Soluble Guanylate Cyclase Stimulator, in Participants with Pulmonary Hypertension Associated with Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2024; 19:1105-1121. [PMID: 38803412 PMCID: PMC11129706 DOI: 10.2147/copd.s454905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
Purpose This phase 1 study (NCT04370873) evaluated safety and pharmacokinetics/pharmacodynamics (PK/PD) of MK-5475 in participants with pulmonary hypertension associated with COPD (PH-COPD). Methods Eligible participants were 40-80 years old with COPD (FEV1/FVC <0.7; FEV1 >30% predicted) and PH (mean pulmonary arterial pressure ≥25 mmHg). Participants were randomized 2:1 to MK-5475 or placebo via dry-powder inhaler once daily for 7 days in Part 1 (360 µg) or 28 days in Part 2 (380 µg). Safety was assessed by adverse events (AEs) and arterial blood oxygenation. Part-2 participants had pulmonary vascular resistance (PVR; primary PD endpoint) and pulmonary blood volume (PBV; secondary PD endpoint) measured at baseline and Day 28. A non-informative prior was used to calculate posterior probability (PP) that the between-group difference (MK-5475 - placebo) in mean percent reduction from baseline in PVR was less than -15%. Results Nine participants were randomized in Part 1, and 14 participants in Part 2. Median age of participants (86.4% male) was 68.5 years (41-77 years); 95.5% had moderate-to-severe COPD. Incidences of AEs were comparable between MK-5475 and placebo: overall (5/14 [36%] versus 5/8 [63%]), drug-related (1/14 [7%] versus 2/8 [25%]), and serious (1/14 [7%] versus 1/8 [13%]). MK-5475 caused no meaningful changes in arterial blood oxygenation or PBV. MK-5475 versus placebo led to numerical improvements from baseline in PVR (-21.2% [95% CI: -35.4, -7.0] versus -5.4% [95% CI: -83.7, 72.9]), with between-group difference in PVR less than -15% and calculated PP of 51%. Conclusion The favorable safety profile and numerical reductions in PVR observed support further clinical development of inhaled MK-5475 for PH-COPD treatment.
Collapse
Affiliation(s)
| | | | | | - Dan Li
- MRL, Merck & Co., Inc., Rahway, NJ, USA
| | | | - Tom Reynders
- Translational Medicine, MSD Belgium, Brussels, Belgium
| | | | - Vasile Corcea
- PMSI Republican Clinical Hospital “T. Mosneaga”, ARENSIA EM Unit, Chisinau, Republic of Moldova
| | - Ketan P Buch
- Department of Internal Medicine, Pulmonary and Critical Care Medicine, Lexington VA Healthcare, Lexington, KY, USA
| | - Eseng Lai
- MRL, Merck & Co., Inc., Rahway, NJ, USA
| | | |
Collapse
|
3
|
Tanabe N, Kumamaru H, Tamura Y, Kondoh Y, Nakayama K, Kinukawa N, Kimura T, Nishiyama O, Tsujino I, Shigeta A, Morio Y, Inoue Y, Kuraishi H, Hirata KI, Tanaka K, Kuwana M, Nagaoka T, Handa T, Sugimura K, Sakamaki F, Naito A, Taniguchi Y, Matsubara H, Hanaoka M, Inami T, Hayama N, Nishimura Y, Kimura H, Miyata H, Tatsumi K. Pulmonary Hypertension With Interstitial Pneumonia: Initial Treatment Effectiveness and Severity in a Japan Registry. JACC. ASIA 2024; 4:403-417. [PMID: 38765657 PMCID: PMC11099821 DOI: 10.1016/j.jacasi.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 01/04/2024] [Accepted: 01/06/2024] [Indexed: 05/22/2024]
Abstract
Background Recent guidelines discourage the use of pulmonary arterial hypertension (PAH)-targeted therapies in patients with pulmonary hypertension (PH) associated with respiratory diseases. Therefore, stratifications of the effectiveness of PAH-targeted therapies are important for this group. Objectives The authors aimed to identify phenotypes that might benefit from initial PAH-targeted therapies in patients with PH associated with interstitial pneumonia and combined pulmonary fibrosis and emphysema. Methods We categorized 270 patients with precapillary PH (192 interstitial pneumonia, 78 combined pulmonary fibrosis and emphysema) into severe and mild PH using a pulmonary vascular resistance of 5 WU. We investigated the prognostic factors and compared the prognoses of initial (within 2 months after diagnosis) and noninitial treatment groups, as well as responders (improvements in World Health Organization functional class, pulmonary vascular resistance, and 6-minute walk distance) and nonresponders. Results Among 239 treatment-naive patients, 46.0% had severe PH, 51.8% had mild ventilatory impairment (VI), and 40.6% received initial treatment. In the severe PH with mild VI subgroup, the initial treatment group had a favorable prognosis compared with the noninitial treatment group. The response rate in this group was significantly higher than the others (48.2% vs 21.8%, ratio 2.21 [95% CI: 1.17-4.16]). In multivariate analysis, initial treatment was a better prognostic factor for severe PH but not for mild PH. Within the severe PH subgroup, responders had a favorable prognosis. Conclusions This study demonstrated an increased number of responders to initial PAH-targeted therapy, with a favorable prognosis in severe PH cases with mild VI. A survival benefit was not observed in mild PH cases. (Multi-institutional Prospective Registry in Pulmonary Hypertension associated with Respiratory Disease; UMIN000011541).
Collapse
Affiliation(s)
- Nobuhiro Tanabe
- Pulmonary Hypertension Center, Saiseikai Narashino Hospital, Narashino, Chiba, Japan
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiraku Kumamaru
- Department of Healthcare Quality Assessment, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuichi Tamura
- Pulmonary Hypertension Center, International University of Health and Welfare Mita Hospital, Tokyo, Japan
| | - Yasuhiro Kondoh
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Aichi, Japan
| | | | - Naoko Kinukawa
- Department of Healthcare Quality Assessment, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoki Kimura
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Aichi, Japan
| | - Osamu Nishiyama
- Department of Respiratory Medicine and Allergology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Ichizo Tsujino
- Division of Respiratory and Cardiovascular Innovative Research, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Ayako Shigeta
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yoshiteru Morio
- Center for Pulmonary Diseases and Respiratory Disease Division, National Hospital Organization Tokyo National Hospital, Kiyose, Tokyo, Japan
| | - Yoshikazu Inoue
- Clinical Research Center, National Hospital Organization Kinki-Chuo Chest Medical Center, Osaka, Japan
| | - Hiroshi Kuraishi
- Department of Respiratory Medicine, Nagano Red Cross Hospital, Nagano, Nagano, Japan
| | - Ken-ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kensuke Tanaka
- Department of Chest Medicine, Japan Railway Tokyo General Hospital, Tokyo, Japan
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | - Tetsutaro Nagaoka
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomohiro Handa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichiro Sugimura
- Department of Cardiology, International University of Health and Welfare Narita Hospital, Narita, Japan
| | - Fumio Sakamaki
- Division of Respiratory Disease, Department of Medicine, Tokai University Hachioji Hospital, Hchioji, Japan
| | - Akira Naito
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yu Taniguchi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiromi Matsubara
- Department of Cardiology, Okayama Medical Center, Okayama, Japan
| | - Masayuki Hanaoka
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Takumi Inami
- Division of Cardiology Department of Medicine, Kyorin University Hospital, Mitaka, Tokyo, Japan
| | - Naoki Hayama
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yoshihiro Nishimura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroshi Kimura
- Department of Respiratory Medicine, Fukujuji Hospital, Japan Anti-Tuberculosis Association (JATA), Kiyose, Tokyo, Japan
| | - Hiroaki Miyata
- Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - JRPHS Group
- Pulmonary Hypertension Center, Saiseikai Narashino Hospital, Narashino, Chiba, Japan
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Healthcare Quality Assessment, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Pulmonary Hypertension Center, International University of Health and Welfare Mita Hospital, Tokyo, Japan
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Aichi, Japan
- Department of Cardiovascular Medicine, Shinko Hospital, Kobe, Japan
- Department of Respiratory Medicine and Allergology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
- Division of Respiratory and Cardiovascular Innovative Research, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Center for Pulmonary Diseases and Respiratory Disease Division, National Hospital Organization Tokyo National Hospital, Kiyose, Tokyo, Japan
- Clinical Research Center, National Hospital Organization Kinki-Chuo Chest Medical Center, Osaka, Japan
- Department of Respiratory Medicine, Nagano Red Cross Hospital, Nagano, Nagano, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Department of Chest Medicine, Japan Railway Tokyo General Hospital, Tokyo, Japan
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Cardiology, International University of Health and Welfare Narita Hospital, Narita, Japan
- Division of Respiratory Disease, Department of Medicine, Tokai University Hachioji Hospital, Hchioji, Japan
- Department of Cardiology, Okayama Medical Center, Okayama, Japan
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Division of Cardiology Department of Medicine, Kyorin University Hospital, Mitaka, Tokyo, Japan
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Department of Respiratory Medicine, Fukujuji Hospital, Japan Anti-Tuberculosis Association (JATA), Kiyose, Tokyo, Japan
- Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
4
|
Alqarni AA, Aldhahir AM, Alghamdi SA, Alqahtani JS, Siraj RA, Alwafi H, AlGarni AA, Majrshi MS, Alshehri SM, Pang L. Role of prostanoids, nitric oxide and endothelin pathways in pulmonary hypertension due to COPD. Front Med (Lausanne) 2023; 10:1275684. [PMID: 37881627 PMCID: PMC10597708 DOI: 10.3389/fmed.2023.1275684] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/19/2023] [Indexed: 10/27/2023] Open
Abstract
Pulmonary hypertension (PH) due to chronic obstructive pulmonary disease (COPD) is classified as Group 3 PH, with no current proven targeted therapies. Studies suggest that cigarette smoke, the most risk factor for COPD can cause vascular remodelling and eventually PH as a result of dysfunction and proliferation of pulmonary artery smooth muscle cells (PASMCs) and pulmonary artery endothelial cells (PAECs). In addition, hypoxia is a known driver of pulmonary vascular remodelling in COPD, and it is also thought that the presence of hypoxia in patients with COPD may further exaggerate cigarette smoke-induced vascular remodelling; however, the underlying cause is not fully understood. Three main pathways (prostanoids, nitric oxide and endothelin) are currently used as a therapeutic target for the treatment of patients with different groups of PH. However, drugs targeting these three pathways are not approved for patients with COPD-associated PH due to lack of evidence. Thus, this review aims to shed light on the role of impaired prostanoids, nitric oxide and endothelin pathways in cigarette smoke- and hypoxia-induced pulmonary vascular remodelling and also discusses the potential of using these pathways as therapeutic target for patients with PH secondary to COPD.
Collapse
Affiliation(s)
- Abdullah A. Alqarni
- Department of Respiratory Therapy, Faculty of Medical Rehabilitation Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Respiratory Therapy Unit, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Abdulelah M. Aldhahir
- Respiratory Therapy Department, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Sara A. Alghamdi
- Respiratory Care Department, Al Murjan Hospital, Jeddah, Saudi Arabia
| | - Jaber S. Alqahtani
- Department of Respiratory Care, Prince Sultan Military College of Health Sciences, Dammam, Saudi Arabia
| | - Rayan A. Siraj
- Department of Respiratory Care, College of Applied Medical Sciences, King Faisal University, Al Ahsa, Saudi Arabia
| | - Hassan Alwafi
- Faculty of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Abdulkareem A. AlGarni
- King Abdulaziz Hospital, The Ministry of National Guard Health Affairs, Al Ahsa, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, College of Applied Medical Sciences, Al Ahsa, Saudi Arabia
| | - Mansour S. Majrshi
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Respiratory Medicine, Royal Brompton Hospital, London, United Kingdom
| | - Saad M. Alshehri
- Department of Respiratory Therapy, King Fahad General Hospital, Jeddah, Saudi Arabia
| | - Linhua Pang
- Respiratory Medicine Research Group, Academic Unit for Translational Medical Sciences, University of Nottingham School of Medicine, Nottingham, United Kingdom
| |
Collapse
|
5
|
Upadhyay P, Wu CW, Pham A, Zeki AA, Royer CM, Kodavanti UP, Takeuchi M, Bayram H, Pinkerton KE. Animal models and mechanisms of tobacco smoke-induced chronic obstructive pulmonary disease (COPD). JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2023; 26:275-305. [PMID: 37183431 PMCID: PMC10718174 DOI: 10.1080/10937404.2023.2208886] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide, and its global health burden is increasing. COPD is characterized by emphysema, mucus hypersecretion, and persistent lung inflammation, and clinically by chronic airflow obstruction and symptoms of dyspnea, cough, and fatigue in patients. A cluster of pathologies including chronic bronchitis, emphysema, asthma, and cardiovascular disease in the form of hypertension and atherosclerosis variably coexist in COPD patients. Underlying causes for COPD include primarily tobacco use but may also be driven by exposure to air pollutants, biomass burning, and workplace related fumes and chemicals. While no single animal model might mimic all features of human COPD, a wide variety of published models have collectively helped to improve our understanding of disease processes involved in the genesis and persistence of COPD. In this review, the pathogenesis and associated risk factors of COPD are examined in different mammalian models of the disease. Each animal model included in this review is exclusively created by tobacco smoke (TS) exposure. As animal models continue to aid in defining the pathobiological mechanisms of and possible novel therapeutic interventions for COPD, the advantages and disadvantages of each animal model are discussed.
Collapse
Affiliation(s)
- Priya Upadhyay
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| | - Ching-Wen Wu
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| | - Alexa Pham
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| | - Amir A. Zeki
- Department of Internal Medicine; Division of Pulmonary, Critical Care, and Sleep Medicine, Center for Comparative Respiratory Biology and Medicine, School of Medicine; University of California, Davis, School of Medicine; U.C. Davis Lung Center; Davis, CA USA
| | - Christopher M. Royer
- California National Primate Research Center, University of California, Davis, Davis, CA 95616 USA
| | - Urmila P. Kodavanti
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Minoru Takeuchi
- Department of Animal Medical Science, Kyoto Sangyo University, Kyoto, Japan
| | - Hasan Bayram
- Koc University Research Center for Translational Medicine (KUTTAM), School of Medicine, Istanbul, Turkey
| | - Kent E. Pinkerton
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| |
Collapse
|
6
|
Feng X, Yang C, Sun Z, Kan W, He X, Chen Y, Tuo Y. Risk factors for mortality in patients with acute exacerbation of cor pulmonale in plateau. BMC Pulm Med 2023; 23:238. [PMID: 37400818 PMCID: PMC10318768 DOI: 10.1186/s12890-023-02509-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/03/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND The risk factors for mortality might differ between patients with acute exacerbation of chronic pulmonary heart disease in plains and plateaus, while there is a lack of evidence. METHOD Patients diagnosed with cor pulmonale at Qinghai Provincial People's Hospital were retrospectively included between January 2012 and December 2021. The symptoms, physical and laboratory examination findings, and treatments were collected. Based on the survival within 50 days, we divided the patients into survival and death groups. RESULTS After 1:10 matching according to gender, age, and altitude, 673 patients were included in the study, 69 of whom died. The multivariable Cox proportional hazards analysis showed that NYHA class IV (HR = 2.03, 95%CI: 1.21-3.40, P = 0.007), type II respiratory failure (HR = 3.57, 95%CI: 1.60-7.99, P = 0.002), acid-base imbalance (HR = 1.82, 95%CI: 1.06-3.14, P = 0.031), C-reactive protein (HR = 1.04, 95%CI: 1.01-1.08, P = 0.026), and D-dimer (HR = 1.07, 95%CI: 1.01-1.13, P = 0.014) were risk factors for death in patients with cor pulmonale at high altitude. Among patients living below 2500 m, cardiac injury was a risk factor for death (HR = 2.47, 95%CI: 1.28-4.77, P = 0.007), while no significant association was observed at ≥ 2500 m (P = 0.057). On the contrary, the increase of D-dimer was only a risk factor for the death of patients living 2500 m and above (HR = 1.23, 95% CI: 1.07-1.40, P = 0.003). CONCLUSION NYHA class IV, type II respiratory failure, acid-base imbalance, and C- reactive protein may increase the risk of death in patients with cor pulmonale. Altitude modified the association between cardiac injury, D-dimer, and death in patients with cor pulmonale.
Collapse
Affiliation(s)
- Xiaokai Feng
- Department of Respiratory and Critical Care Medicine, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining, 810007, Qinghai Province, China
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Chenlu Yang
- Department of Epidemiology and Biostatistics, School of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zerui Sun
- Department of Respiratory and Critical Care Medicine, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining, 810007, Qinghai Province, China
| | - Wanrong Kan
- Department of Respiratory and Critical Care Medicine, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining, 810007, Qinghai Province, China
| | - Xiang He
- Department of Respiratory and Critical Care Medicine, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining, 810007, Qinghai Province, China
| | - Yongxin Chen
- Department of Geratology, Qinghai Red Cross Hospital, 55 South Street, Xining, 810000, Qinghai Province, China.
| | - Yajun Tuo
- Department of Respiratory and Critical Care Medicine, Qinghai Provincial People's Hospital, 2 Gonghe Road, Xining, 810007, Qinghai Province, China.
| |
Collapse
|
7
|
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
|
8
|
Goel K, Egersdorf N, Gill A, Cao D, Collum SD, Jyothula SS, Huang HJ, Sauler M, Lee PJ, Majka S, Karmouty-Quintana H, Petrache I. Characterization of pulmonary vascular remodeling and MicroRNA-126-targets in COPD-pulmonary hypertension. Respir Res 2022; 23:349. [PMID: 36522710 PMCID: PMC9756782 DOI: 10.1186/s12931-022-02267-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Despite causing increased morbidity and mortality, pulmonary hypertension (PH) in chronic obstructive pulmonary disease (COPD) patients (COPD-PH) lacks treatment, due to incomplete understanding of its pathogenesis. Hypertrophy of pulmonary arterial walls and pruning of the microvasculature with loss of capillary beds are known features of pulmonary vascular remodeling in COPD. The remodeling features of pulmonary medium- and smaller vessels in COPD-PH lungs are less well described and may be linked to maladaptation of endothelial cells to chronic cigarette smoking (CS). MicroRNA-126 (miR126), a master regulator of endothelial cell fate, has divergent functions that are vessel-size specific, supporting the survival of large vessel endothelial cells and inhibiting the proliferation of microvascular endothelial cells. Since CS decreases miR126 in microvascular lung endothelial cells, we set out to characterize the remodeling by pulmonary vascular size in COPD-PH and its relationship with miR126 in COPD and COPD-PH lungs. METHODS Deidentified lung tissue was obtained from individuals with COPD with and without PH and from non-diseased non-smokers and smokers. Pulmonary artery remodeling was assessed by ⍺-smooth muscle actin (SMA) abundance via immunohistochemistry and analyzed by pulmonary artery size. miR126 and miR126-target abundance were quantified by qPCR. The expression levels of ceramide, ADAM9, and endothelial cell marker CD31 were assessed by immunofluorescence. RESULTS Pulmonary arteries from COPD and COPD-PH lungs had significantly increased SMA abundance compared to non-COPD lungs, especially in small pulmonary arteries and the lung microvasculature. This was accompanied by significantly fewer endothelial cell markers and increased pro-apoptotic ceramide abundance. miR126 expression was significantly decreased in lungs of COPD individuals. Of the targets tested (SPRED1, VEGF, LAT1, ADAM9), lung miR126 most significantly inversely correlated with ADAM9 expression. Compared to controls, ADAM9 was significantly increased in COPD and COPD-PH lungs, predominantly in small pulmonary arteries and lung microvasculature. CONCLUSION Both COPD and COPD-PH lungs exhibited significant remodeling of the pulmonary vascular bed of small and microvascular size, suggesting these changes may occur before or independent of the clinical development of PH. Decreased miR126 expression with reciprocal increase in ADAM9 may regulate endothelial cell survival and vascular remodeling in small pulmonary arteries and lung microvasculature in COPD and COPD-PH.
Collapse
Affiliation(s)
- Khushboo Goel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care, University of Colorado, Aurora, USA
| | - Nicholas Egersdorf
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, USA
| | - Amar Gill
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, USA
- Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Danting Cao
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, USA
| | - Scott D Collum
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center Houston, Houston, USA
| | - Soma S Jyothula
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, USA
| | - Howard J Huang
- Division of Pulmonary Critical Care, Transplant Pulmonology, Houston Methodist Hospital, Houston, USA
| | - Maor Sauler
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Yale School of Medicine , New Haven, USA
| | - Patty J Lee
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University School of Medicine, Durham, USA
| | - Susan Majka
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care, University of Colorado, Aurora, USA
| | - Harry Karmouty-Quintana
- Divisions of Critical Care, Pulmonary and Sleep Medicine, Department of Internal Medicine, and Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, USA
| | - Irina Petrache
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, USA.
- Department of Medicine, Division of Pulmonary Sciences and Critical Care, University of Colorado, Aurora, USA.
| |
Collapse
|
9
|
Suraya R, Nagano T, Ryanto GRT, Effendi WI, Hazama D, Katsurada N, Yamamoto M, Tachihara M, Emoto N, Nishimura Y, Kobayashi K. Budesonide/glycopyrronium/formoterol fumarate triple therapy prevents pulmonary hypertension in a COPD mouse model via NFκB inactivation. Respir Res 2022; 23:173. [PMID: 35761394 PMCID: PMC9238100 DOI: 10.1186/s12931-022-02081-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/07/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a health problem that results in death, commonly due to the development of pulmonary hypertension (PH). Here, by utilizing a mouse model of intratracheal elastase-induced emphysema that presents three different phases of COPD, we sought to observe whether budesonide/glycopyrronium/formoterol fumarate (BGF) triple therapy could prevent COPD-PH in addition to ameliorating COPD progression. METHODS We utilized intratracheal elastase-induced emphysema mouse model and performed experiments in three phases illustrating COPD progression: inflammatory (1 day post-elastase), emphysema (3 weeks post-elastase) and PH (4 weeks post-elastase), while treatments of BGF and controls (vehicle, one-drug, and two-drug combinations) were started in prior to elastase instillation (inflammatory phase), at day 7 (emphysema), or at day 14 (PH phase). Phenotype analyses were performed in each phase. In vitro, A549 cells or isolated mouse lung endothelial cells (MLEC) were treated with TNFα with/without BGF treatment to analyze NFκB signaling and cytokine expression changes. RESULTS We observed significant reductions in the proinflammatory phenotype observed in the lungs and bronchoalveolar lavage fluid (BALF) 1 day after elastase administration in mice treated with BGF compared with that in mice administered elastase alone (BALF neutrophil percentage, p = 0.0011 for PBS/Vehicle vs. PBS/Elastase, p = 0.0161 for PBS/Elastase vs. BGF). In contrast, only BGF treatment significantly ameliorated the elastase-induced emphysematous lung structure and desaturation after three weeks of elastase instillation (mean linear intercept, p = 0.0156 for PBS/Vehicle vs. PBS/Elastase, p = 0.0274 for PBS/Elastase vs. BGF). Furthermore, BGF treatment prevented COPD-PH development, as shown by improvements in the hemodynamic and histological phenotypes four weeks after elastase treatment (right ventricular systolic pressure, p = 0.0062 for PBS/Vehicle vs. PBS/Elastase, p = 0.027 for PBS/Elastase vs. BGF). Molecularly, BGF acts by inhibiting NFκB-p65 phosphorylation and subsequently decreasing the mRNA expression of proinflammatory cytokines in both alveolar epithelial and pulmonary endothelial cells. CONCLUSION Our results collectively showed that BGF treatment could prevent PH in addition to ameliorating COPD progression via the inhibition of inflammatory NFκB signaling.
Collapse
Affiliation(s)
- Ratoe Suraya
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan.
| | - Gusty Rizky Teguh Ryanto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyama Kitamachi, Higashinada, Kobe, Japan
| | - Wiwin Is Effendi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Daisuke Hazama
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Naoko Katsurada
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Masatsugu Yamamoto
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Motoko Tachihara
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Noriaki Emoto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyama Kitamachi, Higashinada, Kobe, Japan.,Division of Cardiovascular Medicine, Department of Internal Medicine,, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Yoshihiro Nishimura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Kazuyuki Kobayashi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| |
Collapse
|
10
|
Alqarni AA, Brand OJ, Pasini A, Alahmari M, Alghamdi A, Pang L. Imbalanced prostanoid release mediates cigarette smoke-induced human pulmonary artery cell proliferation. Respir Res 2022; 23:136. [PMID: 35643499 PMCID: PMC9145181 DOI: 10.1186/s12931-022-02056-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 05/10/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Pulmonary hypertension is a common and serious complication of chronic obstructive pulmonary disease (COPD). Studies suggest that cigarette smoke can initiate pulmonary vascular remodelling by stimulating cell proliferation; however, the underlying cause, particularly the role of vasoactive prostanoids, is unclear. We hypothesize that cigarette smoke extract (CSE) can induce imbalanced vasoactive prostanoid release by differentially modulating the expression of respective synthase genes in human pulmonary artery smooth muscle cells (PASMCs) and endothelial cells (PAECs), thereby contributing to cell proliferation. METHODS Aqueous CSE was prepared from 3R4F research-grade cigarettes. Human PASMCs and PAECs were treated with or without CSE. Quantitative real-time RT-PCR and Western blotting were used to analyse the mRNA and protein expression of vasoactive prostanoid syhthases. Prostanoid concentration in the medium was measured using ELISA kits. Cell proliferation was assessed using the cell proliferation reagent WST-1. RESULTS We demonstrated that CSE induced the expression of cyclooxygenase-2 (COX-2), the rate-limiting enzyme in prostanoid synthesis, in both cell types. In PASMCs, CSE reduced the downstream prostaglandin (PG) I synthase (PGIS) mRNA and protein expression and PGI2 production, whereas in PAECs, CSE downregulated PGIS mRNA expression, but PGIS protein was undetectable and CSE had no effect on PGI2 production. CSE increased thromboxane (TX) A synthase (TXAS) mRNA expression and TXA2 production, despite undetectable TXAS protein in both cell types. CSE also reduced microsomal PGE synthase-1 (mPGES-1) protein expression and PGE2 production in PASMCs, but increased PGE2 production despite unchanged mPGES-1 protein expression in PAECs. Furthermore, CSE stimulated proliferation of both cell types, which was significantly inhibited by the selective COX-2 inhibitor celecoxib, the PGI2 analogue beraprost and the TXA2 receptor antagonist daltroban. CONCLUSIONS These findings provide the first evidence that cigarette smoke can induce imbalanced prostanoid mediator release characterized by the reduced PGI2/TXA2 ratio and contribute to pulmonary vascular remodelling and suggest that TXA2 may represent a novel therapeutic target for pulmonary hypertension in COPD.
Collapse
Affiliation(s)
- Abdullah A Alqarni
- Respiratory Medicine Research Group, Academic Unit for Translational Medical Sciences, University of Nottingham School of Medicine, City Hospital Campus, Nottingham, NG5 1PB, UK
- Department of Respiratory Therapy, Faculty of Medical Rehabilitation Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Oliver J Brand
- Respiratory Medicine Research Group, Academic Unit for Translational Medical Sciences, University of Nottingham School of Medicine, City Hospital Campus, Nottingham, NG5 1PB, UK
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Alice Pasini
- Respiratory Medicine Research Group, Academic Unit for Translational Medical Sciences, University of Nottingham School of Medicine, City Hospital Campus, Nottingham, NG5 1PB, UK
- Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, Via dell'Università 50, 47522, Cesena, FC, Italy
| | - Mushabbab Alahmari
- Respiratory Medicine Research Group, Academic Unit for Translational Medical Sciences, University of Nottingham School of Medicine, City Hospital Campus, Nottingham, NG5 1PB, UK
- Faculty of Applied Medical Sciences, Department of Respiratory Therapy, University of Bisha, 255, Al Nakhil, Bisha, 67714, Saudi Arabia
| | - Abdulrhman Alghamdi
- Respiratory Medicine Research Group, Academic Unit for Translational Medical Sciences, University of Nottingham School of Medicine, City Hospital Campus, Nottingham, NG5 1PB, UK
- Department of Rehabilitation Science, Respiratory Care Program, King Saud University, Riyadh, Saudi Arabia
| | - Linhua Pang
- Respiratory Medicine Research Group, Academic Unit for Translational Medical Sciences, University of Nottingham School of Medicine, City Hospital Campus, Nottingham, NG5 1PB, UK.
| |
Collapse
|
11
|
Aaronson PI, Rocher A. Oxygen Sensing: Physiology and Pathophysiology. Antioxidants (Basel) 2022; 11:1018. [PMID: 35624881 PMCID: PMC9138106 DOI: 10.3390/antiox11051018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 02/01/2023] Open
Abstract
Oxygen is such an essential element for life that multiple mechanisms have evolved to maintain oxygen homeostasis, including those which detect decreases in arterial O2 and generate adaptive responses to hypoxia [...].
Collapse
Affiliation(s)
- Philip I. Aaronson
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, UK
| | - Asuncion Rocher
- Departamento de Bioquímica, Biología Molecular y Fisiología, Facultad de Medicina, University of Valladolid, 47005 Valladolid, Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), University of Valladolid-CSIC, 47005 Valladolid, Spain
| |
Collapse
|
12
|
Tang M, Wang Y, Wang M, Tong R, Shi T. Risk for Cardiovascular Disease and One-Year Mortality in Patients With Chronic Obstructive Pulmonary Disease and Obstructive Sleep Apnea Syndrome Overlap Syndrome. Front Pharmacol 2021; 12:767982. [PMID: 34764876 PMCID: PMC8576345 DOI: 10.3389/fphar.2021.767982] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/04/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Patients with chronic obstructive pulmonary disease (COPD) and obstructive sleep apnea (OSAS) overlap syndrome (OS) are thought to be at increased risk for cardiovascular diseases. Objective: To evaluate the burden of cardiovascular diseases and long-term outcomes in patients with OS. Methods: This was a retrospective cohort study. The prevalence of cardiovascular diseases and 1-year mortality were compared among patients diagnosed with OS (OS group), COPD alone (COPD group) and OSAS alone (OSAS group), and Cox proportional hazards models were used to assess independent risk factors for all-cause mortality. Results: Overall, patients with OS were at higher risk for pulmonary hypertension (PH), heart failure and all-cause mortality than patients with COPD or OSAS (all p < 0.05). In multivariate Cox regression analysis, the Charlson comorbidity index (CCI) score [adjusted hazard ratio (aHR): 1.273 (1.050–1.543); p = 0.014], hypertension [aHR: 2.006 (1.005–4.004); p = 0.048], pulmonary thromboembolism (PTE) [aHR: 4.774 (1.335–17.079); p = 0.016] and heart failure [aHR: 3.067 (1.521–6.185); p = 0.002] were found to be independent risk factors for 1-year all-cause mortality. Conclusion: Patients with OS had an increased risk for cardiovascular diseases and 1-year mortality. More efforts are needed to identify the causal relationship between OS and cardiovascular diseases, promoting risk stratification and the management of these patients.
Collapse
Affiliation(s)
- Manyun Tang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yidan Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mengjie Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rui Tong
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tao Shi
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
13
|
iTRAQ-based quantitative proteomic analysis of the improved effects of total flavones of Dracocephalum Moldavica L. in chronic mountain sickness. Sci Rep 2021; 11:17526. [PMID: 34471201 PMCID: PMC8410788 DOI: 10.1038/s41598-021-97091-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 08/19/2021] [Indexed: 01/14/2023] Open
Abstract
To use isobaric tags for relative and absolute quantification (iTRAQ) technology to study the pathogenesis of chronic mountain sickness (CMS), identify biomarkers for CMS, and investigate the effect of total flavones of Dracocephalum moldavica L. (TFDM) on a rat model of CMS. We simulated high altitude hypobaric hypoxia conditions and generated a rat model of CMS. Following the administration of TFDM, we measured the pulmonary artery pressure and serum levels of hemoglobin (Hb), the hematocrit (Hct), and observed the structure of the pulmonary artery in experimental rats. Furthermore, we applied iTRAQ-labeled quantitative proteomics technology to identify differentially expressed proteins (DEPs) in the serum, performed bioinformatics analysis, and verified the DEPs by immunohistochemistry. Analysis showed that the pulmonary artery pressure, serum levels of Hb, and the Hct, were significantly increased in a rat model of CMS (P < 0.05). Pathological analysis of lung tissue and pulmonary artery tissue showed that the alveolar compartment had obvious hyperplasia and the pulmonary artery degree of muscularization was enhanced. Both pulmonary artery pressure and tissue morphology were improved following the administration of TFDM. We identified 532 DEPs by quantitative proteomics; gene ontology (GO)and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis further revealed that metabolic pathways associated with coagulation and complement play crucial roles in the occurrence of CMS. Immunohistochemistry verified that several DEPs (α-1-acid glycoprotein, collagen, fibulin, haptoglobin, PLTP, and TAGLN2) are important biological markers for CMS. Our analyses demonstrated that TFDM can improve CMS and exert action by influencing the metabolic pathways associated with coagulation and complement. This process relieves pulmonary artery pressure and improves lung function. We also identified that α-1-acid glycoprotein, collagen, fibulin, haptoglobin, PLTP, and TAGLN2 may represent potential biomarkers for CMS.
Collapse
|
14
|
Abuserewa ST, Selim A, Youssef A, Zolty R. Role of Selexipag in Chronic Obstructive Pulmonary Disease (COPD) Patients With Out-of-Proportion Pulmonary Hypertension. Cureus 2021; 13:e16520. [PMID: 34430131 PMCID: PMC8376144 DOI: 10.7759/cureus.16520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2021] [Indexed: 11/17/2022] Open
Abstract
Pulmonary hypertension (PH) in patients with chronic obstructive pulmonary disease (COPD) is associated with an increase in the risk of COPD exacerbation, increased hospitalization, and worse survival in this patient population. No specific treatment is available for PH in COPD. However, reported out-of-proportion PH may benefit from a certain type of treatment. This study shows that the use of selexipag in the treatment of out-of-proportion PH in COPD patients was associated with an improvement in functional status evaluated by a six-minute walk test (6MWT) and a mean pulmonary artery pressure at 6 +/- 2 months of treatment.
Collapse
Affiliation(s)
- Sherif T Abuserewa
- Internal Medicine, Mercer University School of Medicine, Grand Strand Health, Myrtle Beach, USA
| | - Ahmed Selim
- Department of Cardiovascular Medicine, University of Nebraska Medical Center, Omaha, USA
| | - Amr Youssef
- Department of Cardiovascular Medicine, University of Nebraska Medical Center, Omaha, USA
| | - Ronald Zolty
- Department of Cardiovascular Medicine, University of Nebraska Medical Center, Omaha, USA
| |
Collapse
|
15
|
Sevilla-Montero J, Labrousse-Arias D, Fernández-Pérez C, Fernández-Blanco L, Barreira B, Mondéjar-Parreño G, Alfaro-Arnedo E, López IP, Pérez-Rial S, Peces-Barba G, Pichel JG, Peinado VI, Cogolludo Á, Calzada MJ. Cigarette Smoke Directly Promotes Pulmonary Arterial Remodeling and Kv7.4 Channel Dysfunction. Am J Respir Crit Care Med 2021; 203:1290-1305. [PMID: 33306938 DOI: 10.1164/rccm.201911-2238oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 12/10/2020] [Indexed: 01/10/2023] Open
Abstract
Rationale: Cigarette smoke is considered the chief leading cause of chronic obstructive pulmonary disease (COPD). Its impact on the progressive deterioration of airways has been extensively studied, but its direct effects on the pulmonary vasculature are less known. Objectives: To prove that pulmonary arterial remodeling in patients with COPD is not just a consequence of alveolar hypoxia but also due to the direct effects of cigarette smoke on the pulmonary vascular bed. Methods: We have used different molecular and cell biology approaches, as well as traction force microscopy, wire myography, and patch-clamp techniques in human cells and freshly isolated pulmonary arteries. In addition, we relied on in vivo models and human samples to analyze the effects of cigarette smoke on pulmonary vascular tone alterations. Measurements and Main Results: Cigarette smoke extract exposure directly promoted a hypertrophic, senescent phenotype that in turn contributed, through the secretion of inflammatory molecules, to an increase in the proliferative potential of nonexposed cells. Interestingly, these effects were significantly reversed by antioxidants. Furthermore, cigarette smoke extract affected cell contractility and dysregulated the expression and activity of the voltage-gated K+ channel Kv7.4. This contributed to the impairment of vasoconstriction and vasodilation responses. Most importantly, the levels of this channel were diminished in the lungs of smoke-exposed mice, smokers, and patients with COPD. Conclusions: Cigarette smoke directly contributes to pulmonary arterial remodeling through increased cell senescence, as well as vascular tone alterations because of diminished levels and function in the Kv7.4 channel. Strategies targeting these pathways may lead to novel therapies for COPD.
Collapse
Affiliation(s)
- Javier Sevilla-Montero
- Biomedical Research Institute La Princesa Hospital, Madrid, Spain
- Department of Medicine, School of Medicine, and
- Doctoral School, Autonoma University of Madrid, Madrid, Spain
| | - David Labrousse-Arias
- Biomedical Research Institute La Princesa Hospital, Madrid, Spain
- Department of Medicine, School of Medicine, and
| | - Cintia Fernández-Pérez
- Biomedical Research Institute La Princesa Hospital, Madrid, Spain
- Department of Medicine, School of Medicine, and
| | - Laura Fernández-Blanco
- Biomedical Research Institute La Princesa Hospital, Madrid, Spain
- Department of Medicine, School of Medicine, and
| | - Bianca Barreira
- Department of Pharmacology and Toxicology, School of Medicine, Complutense University of Madrid, Madrid, Spain
- Biomedical Research Networking Center in Respiratory Diseases, Institute of Health Carlos III, Madrid, Spain
| | - Gema Mondéjar-Parreño
- Department of Pharmacology and Toxicology, School of Medicine, Complutense University of Madrid, Madrid, Spain
- Biomedical Research Networking Center in Respiratory Diseases, Institute of Health Carlos III, Madrid, Spain
| | - Elvira Alfaro-Arnedo
- Lung Cancer and Respiratory Diseases Unit, Biomedical Research Center of La Rioja, Rioja Salud Foundation, Logroño, Spain
| | - Icíar P López
- Lung Cancer and Respiratory Diseases Unit, Biomedical Research Center of La Rioja, Rioja Salud Foundation, Logroño, Spain
| | - Sandra Pérez-Rial
- Biomedical Research Networking Center in Respiratory Diseases, Institute of Health Carlos III, Madrid, Spain
- Respiratory Research Unit, Biomedical Research Unit, Health Research Institute Fundación Jiménez Díaz, Madrid, Spain; and
| | - Germán Peces-Barba
- Biomedical Research Networking Center in Respiratory Diseases, Institute of Health Carlos III, Madrid, Spain
- Respiratory Research Unit, Biomedical Research Unit, Health Research Institute Fundación Jiménez Díaz, Madrid, Spain; and
| | - José G Pichel
- Biomedical Research Networking Center in Respiratory Diseases, Institute of Health Carlos III, Madrid, Spain
- Lung Cancer and Respiratory Diseases Unit, Biomedical Research Center of La Rioja, Rioja Salud Foundation, Logroño, Spain
| | - Víctor Ivo Peinado
- Biomedical Research Networking Center in Respiratory Diseases, Institute of Health Carlos III, Madrid, Spain
- Department of Pulmonary Medicine, Hospital August Pi i Sunyer Biomedical Research Institute, University of Barcelona, Barcelona, Spain
| | - Ángel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, Complutense University of Madrid, Madrid, Spain
- Biomedical Research Networking Center in Respiratory Diseases, Institute of Health Carlos III, Madrid, Spain
| | - María J Calzada
- Biomedical Research Institute La Princesa Hospital, Madrid, Spain
- Department of Medicine, School of Medicine, and
| |
Collapse
|
16
|
Wang Y, Li X, Niu W, Chen J, Zhang B, Zhang X, Wang Y, Dang S, Li Z. The alveolar epithelial cells are involved in pulmonary vascular remodeling and constriction of hypoxic pulmonary hypertension. Respir Res 2021; 22:134. [PMID: 33947399 PMCID: PMC8094493 DOI: 10.1186/s12931-021-01708-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/08/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hypoxic pulmonary hypertension (HPH) is a common type of pulmonary hypertension and characterized by pulmonary vascular remodeling and constriction. Alveolar epithelial cells (AECs) primarily sense alveolar hypoxia, but the role of AECs in HPH remains unclear. In this study, we explored whether AECs are involved in pulmonary vascular remodeling and constriction. METHODS In the constructed rat HPH model, hemodynamic and morphological characteristics were measured. By treating AECs with hypoxia, we further detected the levels of superoxide dismutase 2 (SOD2), catalase (CAT), reactive oxygen species (ROS) and hydrogen peroxide (H2O2), respectively. To detect the effects of AECs on pulmonary vascular remodeling and constriction, AECs and pulmonary artery smooth cells (PASMCs) were co-cultured under hypoxia, and PASMCs and isolated pulmonary artery (PA) were treated with AECs hypoxic culture medium. In addition, to explore the mechanism of AECs on pulmonary vascular remodeling and constriction, ROS inhibitor N-acetylcysteine (NAC) was used. RESULTS Hypoxia caused pulmonary vascular remodeling and increased pulmonary artery pressure, but had little effect on non-pulmonary vessels in vivo. Meanwhile, in vitro, hypoxia promoted the imbalance of SOD2 and CAT in AECs, leading to increased ROS and hydrogen peroxide (H2O2) production in the AECs culture medium. In addition, AECs caused the proliferation of co-cultured PASMCs under hypoxia, and the hypoxic culture medium of AECs enhanced the constriction of isolated PA. However, treatment with ROS inhibitor NAC effectively alleviated the above effects. CONCLUSION The findings of present study demonstrated that AECs were involved in pulmonary vascular remodeling and constriction under hypoxia by paracrine H2O2 into the pulmonary vascular microenvironment.
Collapse
Affiliation(s)
- Yanxia Wang
- Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Xiaoming Li
- Department of Pathophysiology, Xi'an Peihua University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Wen Niu
- Department of Pathophysiology, School of Basic Medicine, Fourth Military Medical University, 169 Changle Western Street, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Jian Chen
- Department of Respiratory and Critical Care, Tangdu Hospital, Fourth Military Medical University, 569 Xinsi Street, Xi'an, Shaanxi, 710038, People's Republic of China
| | - Bo Zhang
- Department of Pathophysiology, School of Basic Medicine, Fourth Military Medical University, 169 Changle Western Street, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Xiumin Zhang
- Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Yingmei Wang
- Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Shaokang Dang
- Department of Respiratory and Critical Care, Tangdu Hospital, Fourth Military Medical University, 569 Xinsi Street, Xi'an, Shaanxi, 710038, People's Republic of China.
| | - Zhichao Li
- Department of Pathophysiology, School of Basic Medicine, Fourth Military Medical University, 169 Changle Western Street, Xi'an, Shaanxi, 710032, People's Republic of China.
- Northwest University School of Medicine, Xi'an, Shaanxi, 710075, People's Republic of China.
| |
Collapse
|
17
|
Mandoli GE, De Carli G, Pastore MC, Cameli P, Contorni F, D'Alessandro M, Bargagli E, Mondillo S, Cameli M. Right cardiac involvement in lung diseases: a multimodality approach from diagnosis to prognostication. J Intern Med 2021; 289:440-449. [PMID: 32996153 DOI: 10.1111/joim.13179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/12/2020] [Accepted: 09/17/2020] [Indexed: 02/04/2023]
Abstract
Lung diseases are amongst the main healthcare issues in the general population, having a high burden of morbidity and mortality. The cardiovascular system has a key role in patients affected by respiratory disorders. More specifically, the right ventricle (RV) enables the impaired lung function to be overcome in an initial stage of disease process, reducing the severity of dyspnoea. In addition, two of the main causes of death in this setting are RV failure and sudden cardiac death (SCD). Echocardiography is regarded as a useful and easily available tool in assessing RV function. Several noninvasive echocardiographic parameters of elevated pulmonary pressures and RV function have been proposed. The combination of different parameters and imaging methods is paramount and researches regarding RV impairment using these indices has been specifically addressed in relation to the chronic obstructive and restrictive lung disease in order to guide the clinicians in the management of these patients. Cardiac involvement in lung diseases is often observed, and RV changes are reported also in early stages of pulmonary diseases. The role of right ventricle in chronic respiratory disease patients has to be evaluated in detail to describe the response to therapy and the degree of disease progression through multimodality and advanced imaging techniques. The aim of this review is to describe the different pathophysiological mechanisms of cardiac impairment in primary lung disease (such as chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF) and sarcoidosis) and to summarize the role of cardiac multimodality imaging in the diagnosis and the prognosis of these diseases.
Collapse
Affiliation(s)
- G E Mandoli
- From the, Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
| | - G De Carli
- From the, Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
| | - M C Pastore
- From the, Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
| | - P Cameli
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - F Contorni
- From the, Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
| | - M D'Alessandro
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - E Bargagli
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - S Mondillo
- From the, Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
| | - M Cameli
- From the, Department of Medical Biotechnologies, Division of Cardiology, University of Siena, Siena, Italy
| |
Collapse
|
18
|
Oxygen-sensitivity and Pulmonary Selectivity of Vasodilators as Potential Drugs for Pulmonary Hypertension. Antioxidants (Basel) 2021; 10:antiox10020155. [PMID: 33494520 PMCID: PMC7911835 DOI: 10.3390/antiox10020155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 11/23/2022] Open
Abstract
Current approved therapies for pulmonary hypertension (PH) aim to restore the balance between endothelial mediators in the pulmonary circulation. These drugs may exert vasodilator effects on poorly oxygenated vessels. This may lead to the derivation of blood perfusion towards low ventilated alveoli, i.e., producing ventilation-perfusion mismatch, with detrimental effects on gas exchange. The aim of this study is to analyze the oxygen-sensitivity in vitro of 25 drugs currently used or potentially useful for PH. Additionally, the study analyses the effectiveness of these vasodilators in the pulmonary vs. the systemic vessels. Vasodilator responses were recorded in pulmonary arteries (PA) and mesenteric arteries (MA) from rats and in human PA in a wire myograph under different oxygen concentrations. None of the studied drugs showed oxygen selectivity, being equally or more effective as vasodilators under conditions of low oxygen as compared to high oxygen levels. The drugs studied showed low pulmonary selectivity, being equally or more effective as vasodilators in systemic than in PA. A similar behavior was observed for the members within each drug family. In conclusion, none of the drugs showed optimal vasodilator profile, which may limit their therapeutic efficacy in PH.
Collapse
|
19
|
Urban MH, Mayr AK, Schmidt I, Margulies E, Grasmuk-Siegl E, Burghuber OC, Funk GC. Induction of dynamic hyperinflation by expiratory resistance breathing in healthy subjects - an efficacy and safety study. Exp Physiol 2020; 106:532-543. [PMID: 33174314 PMCID: PMC7894562 DOI: 10.1113/ep088439] [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] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 11/09/2020] [Indexed: 12/19/2022]
Abstract
New Findings What is the central question of this study? The study aimed to establish a novel model to study the chronic obstructive pulmonary disease (COPD)‐related cardiopulmonary effects of dynamic hyperinflation in healthy subjects. What is the main finding and its importance? A model of expiratory resistance breathing (ERB) was established in which dynamic hyperinflation was induced in healthy subjects, expressed both by lung volumes and intrathoracic pressures. ERB outperformed existing methods and represents an efficacious model to study cardiopulmonary mechanics of dynamic hyperinflation without potentially confounding factors as present in COPD.
Abstract Dynamic hyperinflation (DH) determines symptoms and prognosis of chronic obstructive pulmonary disease (COPD). The induction of DH is used to study cardiopulmonary mechanics in healthy subjects without COPD‐related confounders like inflammation, hypoxic vasoconstriction and rarefication of pulmonary vasculature. Metronome‐paced tachypnoea (MPT) has proven effective in inducing DH in healthy subjects, but does not account for airflow limitation. We aimed to establish a novel model incorporating airflow limitation by combining tachypnoea with an expiratory airway stenosis. We investigated this expiratory resistance breathing (ERB) model in 14 healthy subjects using different stenosis diameters to assess a dose–response relationship. Via cross‐over design, we compared ERB to MPT in a random sequence. DH was quantified by inspiratory capacity (IC, litres) and intrinsic positive end‐expiratory pressure (PEEPi, cmH2O). ERB induced a stepwise decreasing IC (means (95% CI): tidal breathing: 3.66 (3.45–3.88), ERB 3 mm: 3.33 (1.75–4.91), 2 mm: 2.05 (0.76–3.34), 1.5 mm: 0.73 (0.12–1.58) litres) and increasing PEEPi (tidal breathing: 0.70 (0.50–0.80), ERB 3 mm: 11.1 (7.0–15.2), 2 mm: 22.3 (17.1–27.6), 1.5 mm: 33.4 (3.40–63) cmH2O). All three MPT patterns increased PEEPi, but to a far lesser extent than ERB. No adverse events during ERB were noted. In conclusion, ERB was proven to be a safe and efficacious model for the induction of DH and might be used for the investigation of cardiopulmonary interaction in healthy subjects.
Collapse
Affiliation(s)
- Matthias Helmut Urban
- Department of Internal and Respiratory Medicine, Krankenhaus Nord - Klinik Floridsdorf, Vienna, Austria.,Karl-Landsteiner-Institute for Lung Research and Pulmonary Oncology, Vienna, Austria.,Ludwig-Boltzmann Institute for COPD and Respiratory Epidemiology, Vienna, Austria
| | - Anna Katharina Mayr
- Department of Internal and Respiratory Medicine, Krankenhaus Nord - Klinik Floridsdorf, Vienna, Austria.,Karl-Landsteiner-Institute for Lung Research and Pulmonary Oncology, Vienna, Austria
| | - Ingrid Schmidt
- Department of Internal and Respiratory Medicine, Krankenhaus Nord - Klinik Floridsdorf, Vienna, Austria
| | | | - Erwin Grasmuk-Siegl
- Department of Internal and Respiratory Medicine, Krankenhaus Nord - Klinik Floridsdorf, Vienna, Austria.,Karl-Landsteiner-Institute for Lung Research and Pulmonary Oncology, Vienna, Austria
| | - Otto Chris Burghuber
- Ludwig-Boltzmann Institute for COPD and Respiratory Epidemiology, Vienna, Austria.,Medical School, Sigmund Freud University, Vienna, Austria
| | - Georg-Christian Funk
- Karl-Landsteiner-Institute for Lung Research and Pulmonary Oncology, Vienna, Austria.,Department of Internal and Respiratory Medicine, Wilhelminenspital, Vienna, Austria
| |
Collapse
|
20
|
Akanji MA, Adeyanju AA, Rotimi D, Adeyemi OS. Nitric Oxide Balance in Health and Diseases: Implications for New Treatment Strategies. Open Biochem J 2020. [DOI: 10.2174/1874091x02014010025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Nitric Oxide (NO) is an essential signaling molecule with diverse physiological functions in humans. The steady-state concentration and site of production of nitric oxide determine its effects in biological systems. The human cells are exposed to both beneficial and harmful effects of NO. These dual effects of NO could depend on its local concentration in the cells. Additionally, the rate of synthesis, translocation, direct interaction with other molecules, and signals contribute to the biochemical and physiological effects of NO. In this review, the biochemical and physiological role of NO, particularly in health and disease as touching on cell signaling, oxidative stress, immunity, as well as cardiovascular protection amongst others, is focused on. Therefore, this review objectively discusses the dual functionality of NO in living cells.
Collapse
|
21
|
Blanco I, Tura-Ceide O, Peinado VI, Barberà JA. Updated Perspectives on Pulmonary Hypertension in COPD. Int J Chron Obstruct Pulmon Dis 2020; 15:1315-1324. [PMID: 32606641 PMCID: PMC7293405 DOI: 10.2147/copd.s211841] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/24/2020] [Indexed: 12/11/2022] Open
Abstract
Pulmonary hypertension (PH) is a frequent and important complication of chronic obstructive pulmonary disease (COPD). It is associated with worse clinical courses with more frequent exacerbation episodes, shorter survival, and greater need of health resources. PH is usually of moderate severity and progresses slowly, without altering right ventricular function in the majority of cases. Nevertheless, a reduced subgroup of patients may present disproportionate PH, with pulmonary artery pressure (PAP) largely exceeding the severity of respiratory impairment. These patients may represent a group with an exaggerated vascular impairment (pulmonary vascular phenotype) to factors that induce PH in COPD or be patients in whom idiopathic pulmonary arterial hypertension (PAH) coexist. The present review addresses the current definition and classification of PH in COPD, the distinction among the different phenotypes of pulmonary vascular disease that might present in COPD patients, and the therapeutic approach to PH in COPD based on the available scientific evidence.
Collapse
Affiliation(s)
- Isabel Blanco
- Department of Pulmonary Medicine, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Victor Ivo Peinado
- Department of Pulmonary Medicine, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Joan Albert Barberà
- Department of Pulmonary Medicine, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| |
Collapse
|
22
|
Gredic M, Blanco I, Kovacs G, Helyes Z, Ferdinandy P, Olschewski H, Barberà JA, Weissmann N. Pulmonary hypertension in chronic obstructive pulmonary disease. Br J Pharmacol 2020; 178:132-151. [PMID: 31976545 DOI: 10.1111/bph.14979] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 12/29/2019] [Accepted: 01/06/2020] [Indexed: 12/12/2022] Open
Abstract
Even mild pulmonary hypertension (PH) is associated with increased mortality and morbidity in patients with chronic obstructive pulmonary disease (COPD). However, the underlying mechanisms remain elusive; therefore, specific and efficient treatment options are not available. Therapeutic approaches tested in the clinical setting, including long-term oxygen administration and systemic vasodilators, gave disappointing results and might be only beneficial for specific subgroups of patients. Preclinical studies identified several therapeutic approaches for the treatment of PH in COPD. Further research should provide deeper insight into the complex pathophysiological mechanisms driving vascular alterations in COPD, especially as such vascular (molecular) alterations have been previously suggested to affect COPD development. This review summarizes the current understanding of the pathophysiology of PH in COPD and gives an overview of the available treatment options and recent advances in preclinical studies. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.1/issuetoc.
Collapse
Affiliation(s)
- Marija Gredic
- Cardio-Pulmonary Institute, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Isabel Blanco
- Department of Pulmonary Medicine, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Gabor Kovacs
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria.,Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School & János Szentágothai Research Centre, University of Pécs, Pécs, Hungary.,PharmInVivo Ltd, Pécs, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Horst Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria.,Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Joan Albert Barberà
- Department of Pulmonary Medicine, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Norbert Weissmann
- Cardio-Pulmonary Institute, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
23
|
Wang G, Shang W, Ren Y, Liu S, Ren X, Wei S, Han D. Benefits of statins in chronic obstructive pulmonary disease patients with pulmonary hypertension: A meta-analysis. Eur J Intern Med 2019; 70:39-42. [PMID: 31679886 DOI: 10.1016/j.ejim.2019.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 08/29/2019] [Accepted: 09/14/2019] [Indexed: 11/28/2022]
Abstract
PURPOSE This meta-analysis was performed to evaluate the efficacy of statins in chronic obstructive pulmonary disease (COPD) patients with pulmonary hypertension (PH). METHODS A systematic search was made of MEDLINE, Cochrane, ISI Web of Science and SCOPUS databases. Randomized clinical trials on treatment of COPD-PH with the statins, compared with placebo, were reviewed. Studies were pooled to weighted mean differences (WMD), with 95% confidence interval (CI). RESULTS Five trials (enrolling 270 participants) met the inclusion criteria. Compared with placebo, the statins presented significant effects on systolic pulmonary artery pressure (WMD -4.52 mmHg; 95% CI -6.32 to -2.72 mmHg) and 6-min walk distance (6MWD) (WMD 32.46 m; 95% CI 13.63-51.29 m). CONCLUSIONS Statins therapy significantly improves PH and 6MWD in COPD patients with PH.
Collapse
Affiliation(s)
- Guizuo Wang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, Shaanxi 710068, PR China
| | - Wenli Shang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, Shaanxi 710068, PR China
| | - Yajuan Ren
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, Shaanxi 710068, PR China
| | - Sining Liu
- Department of General Medicine, Xi'an Medical University, Xi'an, Shaanxi 710068, PR China
| | - Xiaoping Ren
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, Shaanxi 710068, PR China
| | - Shenghong Wei
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, Shaanxi 710068, PR China
| | - Dong Han
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, No. 256, West Youyi Road, Xi'an, Shaanxi 710068, PR China.
| |
Collapse
|
24
|
Kovacs G, Agusti A, Barberà JA, Celli B, Criner G, Humbert M, Sin DD, Voelkel N, Olschewski H. Pulmonary Vascular Involvement in Chronic Obstructive Pulmonary Disease. Is There a Pulmonary Vascular Phenotype? Am J Respir Crit Care Med 2019; 198:1000-1011. [PMID: 29746142 DOI: 10.1164/rccm.201801-0095pp] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Gabor Kovacs
- 1 Medical University of Graz, Graz, Austria.,2 Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Alvar Agusti
- 3 Respiratory Institute, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, University of Barcelona, Barcelona, Spain.,4 Centro Investigacion Biomedica en Red de Enfermedades Respiratorias, Madrid, Spain
| | - Joan Albert Barberà
- 3 Respiratory Institute, Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, University of Barcelona, Barcelona, Spain.,4 Centro Investigacion Biomedica en Red de Enfermedades Respiratorias, Madrid, Spain
| | | | - Gerard Criner
- 6 Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Marc Humbert
- 7 Université Paris-Sud, Université Paris-Saclay; Inserm U999; Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Don D Sin
- 8 Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada.,9 Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia; Canada; and
| | - Norbert Voelkel
- 10 Department of Pulmonary Medicine, Frije University, Medical Center, Amsterdam, the Netherlands
| | - Horst Olschewski
- 1 Medical University of Graz, Graz, Austria.,2 Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| |
Collapse
|
25
|
Awerbach JD, Stackhouse KA, Lee J, Dahhan T, Parikh KS, Krasuski RA. Outcomes of lung disease-associated pulmonary hypertension and impact of elevated pulmonary vascular resistance. Respir Med 2019; 150:126-130. [PMID: 30961938 DOI: 10.1016/j.rmed.2019.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 11/17/2022]
Abstract
BACKGROUND The clinical characteristics, hemodynamic changes and outcomes of lung disease-associated pulmonary hypertension (LD-PH) are poorly defined. METHODS A prospective cohort of PH patients undergoing initial hemodynamic assessment was collected, from which 51 patients with LD-PH were identified. Baseline characteristics and long-term survival were compared with 83 patients with idiopathic pulmonary arterial hypertension (iPAH). RESULTS Mean age (±standard deviation) of LD-PH patients was 64 ± 10 years, 30% were female and 78% were New York Heart Association class III-IV. The LD-PH group was older than the iPAH group (64 ± 10 vs 56 ± 18 years, respectively, P = 0.003) with a lower percentage of women (30% vs 70%, P = 0.007). LD-PH patients had smaller right ventricular sizes (P = 0.02) and less tricuspid regurgitation (P = 0.03) by echocardiogram, and lower mean pulmonary arterial pressures (mPAP) (P = 0.01) and pulmonary vascular resistance (PVR) (P = 0.001) at catheterization. Despite these findings, mortality was equally high in both groups (P = 0.16). 5-year survival was lower in patients with interstitial lung disease compared to those with obstructive pulmonary disease (P = 0.05). Among the LD-PH population, those with mild to moderately elevated mPAP and those with PVR <7 Wood units demonstrated significantly improved survival (P = 0.04 and P = 0.001, respectively). Vasoreactivity was not associated with improved survival (P = 0.64). A PVR ≥7 Wood units was associated with increased risk of mortality (hazard ratio (95% confidence interval), 3.59 (1.27-10.19), P = 0.02). CONCLUSIONS Despite less severe PH and less right heart sequelae, LD-PH has an equally poor clinical outcome when compared to iPAH. A PVR ≥7 Wood units in LD-PH patients was associated with 3-fold higher mortality.
Collapse
Affiliation(s)
- Jordan D Awerbach
- Duke University Medical Center, Division of Cardiovascular Medicine, Durham, NC, USA
| | | | - Joanne Lee
- The Cleveland Clinic, Cleveland, OH, USA
| | - Talal Dahhan
- Duke University Medical Center, Division of Pulmonary and Critical Care Medicine, Durham, NC, USA
| | - Kishan S Parikh
- Duke University Medical Center, Division of Cardiovascular Medicine, Durham, NC, USA
| | - Richard A Krasuski
- Duke University Medical Center, Division of Cardiovascular Medicine, Durham, NC, USA.
| |
Collapse
|
26
|
Nathan SD, Barbera JA, Gaine SP, Harari S, Martinez FJ, Olschewski H, Olsson KM, Peacock AJ, Pepke-Zaba J, Provencher S, Weissmann N, Seeger W. Pulmonary hypertension in chronic lung disease and hypoxia. Eur Respir J 2019; 53:13993003.01914-2018. [PMID: 30545980 PMCID: PMC6351338 DOI: 10.1183/13993003.01914-2018] [Citation(s) in RCA: 448] [Impact Index Per Article: 74.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 10/09/2018] [Indexed: 02/07/2023]
Abstract
Pulmonary hypertension (PH) frequently complicates the course of patients with various forms of chronic lung disease (CLD). CLD-associated PH (CLD-PH) is invariably associated with reduced functional ability, impaired quality of life, greater oxygen requirements and an increased risk of mortality. The aetiology of CLD-PH is complex and multifactorial, with differences in the pathogenic sequelae between the diverse forms of CLD. Haemodynamic evaluation of PH severity should be contextualised within the extent of the underlying lung disease, which is best gauged through a combination of physiological and imaging assessment. Who, when, if and how to screen for PH will be addressed in this article, as will the current state of knowledge with regard to the role of treatment with pulmonary vasoactive agents. Although such therapy cannot be endorsed given the current state of findings, future studies in this area are strongly encouraged. State of the art and research perspectives in pulmonary hypertension in chronic lung disease and hypoxiahttp://ow.ly/XcW730meWxy
Collapse
Affiliation(s)
| | - Joan A Barbera
- Dept of Pulmonary Medicine, Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Center on Respiratory Diseases, Madrid, Spain
| | - Sean P Gaine
- Respiratory Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Sergio Harari
- U.O. di Pneumologia e Terapia Semi-Intensiva Respiratoria, Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, Ospedale San Giuseppe, MultiMedica IRCCS, Milan, Italy
| | | | - Horst Olschewski
- Division of Pulmonology, Medizinische Universitat Graz, Graz, Austria
| | - Karen M Olsson
- Dept of Respiratory Medicine, Hannover Medical School and Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Andrew J Peacock
- Scottish Pulmonary Vascular Unit, Regional Lung and Heart Centre, Glasgow, UK
| | | | - Steeve Provencher
- Institut Universitaire de Cardiologie et de Pneumologie de Québec Research Center, Laval University, Quebec City, QC, Canada
| | - Norbert Weissmann
- University of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig University Giessen and Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Werner Seeger
- University of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig University Giessen and Member of the German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
27
|
Nashat H, Gatzoulis MA. The importance of left heart disease as a cause of pulmonary hypertension in COPD. Hellenic J Cardiol 2018; 59:166-167. [PMID: 29940220 DOI: 10.1016/j.hjc.2018.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/01/2018] [Accepted: 06/08/2018] [Indexed: 11/27/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common respiratory condition that presents with varying degrees of severity and can be complicated by further comorbidities. Up to a third can also have pulmonary hypertension, which is an important risk factor associated with an increase in morbidity and mortality. The etiology of pulmonary hypertension contributes to diagnosis. Considering and identifying the type of pulmonary hypertension at the right time will influence management and outcome in patients with COPD.
Collapse
Affiliation(s)
- H Nashat
- Royal Brompton and Harfield Foundation Trust, London, UK; National Heart and Lung Institute, Imperial College London, UK.
| | - M A Gatzoulis
- Royal Brompton and Harfield Foundation Trust, London, UK; National Heart and Lung Institute, Imperial College London, UK
| |
Collapse
|
28
|
Discovery and development of sGC stimulators for the treatment of pulmonary hypertension and rare diseases. Nitric Oxide 2018; 77:88-95. [PMID: 29738821 DOI: 10.1016/j.niox.2018.05.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 12/26/2022]
Abstract
The NO/sGC/cGMP signaling cascade plays a pivotal role in regulation of cardiovascular, cardiopulmonary and cardiorenal diseases and impairment of this cascade results in severe pathologies. Therefore, pharmacological interventions, targeting this pathway are promising strategies for treating a variety of diseases. Nitrates, supplementing NO and, PDE5 inhibitors preventing cGMP degradation, are used for angina pectoris treatment and the treatment of pulmonary arterial hypertension (PAH), respectively. More recently, a new class of drugs which directly stimulate the sGC enzyme and trigger NO-independent cGMP production was introduced and termed sGC stimulators. In 2013, the first sGC stimulator, riociguat, was approved for the treatment of PAH and chronic thromboembolic pulmonary hypertension (CTEPH). Since cGMP targets multiple intracellular downstream targets, sGC stimulators have shown - beyond the well characterized vasodilatation - anti-fibrotic, anti-inflammatory and anti-proliferative effects. These additional modes of action might extend the therapeutic potential of this drug class substantially. This review summarizes the NO/sGC/cGMP signaling cascades, the discovery and the mode of action of sGC stimulators. Furthermore, the preclinical evidence and development of riociguat for the treatment of PAH and CTEPH is reviewed. Finally, a summary of the antifibrotic effects of sGC stimulators, especially the most recent finding for skin fibrosis are included which may indicate efficacy in fibrotic diseases like Systemic Sclerosis (SSc).
Collapse
|
29
|
Nydegger C, Martinelli C, Di Marco F, Bulfamante G, von Segesser L, Tozzi P, Samaja M, Milano G. Phosphodiesterase-5 Inhibition Alleviates Pulmonary Hypertension and Basal Lamina Thickening in Rats Challenged by Chronic Hypoxia. Front Physiol 2018; 9:289. [PMID: 29636700 PMCID: PMC5880920 DOI: 10.3389/fphys.2018.00289] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/12/2018] [Indexed: 11/30/2022] Open
Abstract
Background: Hypoxia represents both an outcome of cardiopulmonary diseases and a trigger for severe pulmonary complications as pulmonary hypertension. Because nitric oxide (NO) is a critical mediator in the development of pulmonary hypertension, the modulators of its downstream function may become target of pharmacological interventions aimed at alleviating the impact of this condition. Here, we investigate the effects of an early administration of phosphodiesterase-5 inhibitor in rats where pulmonary artery hypertension was induced by chronic exposure to hypoxia. Methods: Rats were divided into three groups: normoxic control, hypoxic with no treatments (2 weeks breathing an atmosphere containing 10% oxygen), and hypoxic treated with sildenafil (1.4 mg/Kg per day in 0.3 mL i.p.). After sacrifice, hearts and lungs were removed and harvested for analyses. Results: Sildenafil reduced hypoxia-induced right ventricle hypertrophy without effects in lung hypertrophy, and blunted the increase in right ventricle pressure without effects on left ventricle pressure. Furthermore, the NO-producing systems (i.e., the phosphorylation of the endothelial isoforms of NO synthase that was measured in both myocardial and lung tissues), and the blood NO stores (i.e., the plasma level of nitrates and nitrites) were up-regulated by sildenafil. We did not find significant effects of sildenafil on weight and hemoglobin concentration. Morphological analysis in lung biopsies revealed that 2-week hypoxia increased the frequency of small pulmonary vessels leaving large vessels unaffected. Finally, ultrastructural analysis showed that sildenafil down-regulated the hypoxia-induced increase in the thickness of the pulmonary basal lamina. Conclusions: In this model of pulmonary hypertension, sildenafil contrasts the negative effects of hypoxia on pulmonary vascular and right ventricle remodeling. This action does not only encompass the canonical vasomodulatory effect, but involves several biochemical pathways. Although the human pathological model is certainly more complex than that described here (for example, the inflammatory issue), the potential role of phosphodiesterase-5 for long-term treatment, and perhaps prevention, of pulmonary hypertension is worthy of investigation.
Collapse
Affiliation(s)
- Coline Nydegger
- Laboratory of Cardiovascular Research, Department of Surgery and Anesthesiology, University Hospital of Lausanne, Lausanne, Switzerland
| | | | - Fabiano Di Marco
- Department of Health Science, University of Milan, Milan, Italy.,ASST Santi Paolo e Carlo, Milan, Italy
| | - Gaetano Bulfamante
- Department of Health Science, University of Milan, Milan, Italy.,ASST Santi Paolo e Carlo, Milan, Italy
| | - Ludwig von Segesser
- Laboratory of Cardiovascular Research, Department of Surgery and Anesthesiology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Piergiorgio Tozzi
- Laboratory of Cardiovascular Research, Department of Surgery and Anesthesiology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Michele Samaja
- Department of Health Science, University of Milan, Milan, Italy
| | - Giuseppina Milano
- Laboratory of Cardiovascular Research, Department of Surgery and Anesthesiology, University Hospital of Lausanne, Lausanne, Switzerland
| |
Collapse
|
30
|
Wang L, Jin YZ, Zhao QH, Jiang R, Wu WH, Gong SG, He J, Liu JM, Jing ZC. Hemodynamic and gas exchange effects of inhaled iloprost in patients with COPD and pulmonary hypertension. Int J Chron Obstruct Pulmon Dis 2017; 12:3353-3360. [PMID: 29200842 PMCID: PMC5702173 DOI: 10.2147/copd.s141798] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Studies have shown that vasodilators such as iloprost can be useful for treating pulmonary hypertension (PH). However, in patients with COPD, vasodilators may inhibit hypoxic pulmonary vasoconstriction and impair gas exchange. The efficacy and safety of iloprost inhalation was assessed in 67 patients with PH associated with COPD (COPD-PH), diagnosed by right heart catheterization. Of these, 37 patients had severe PH (mean pulmonary arterial pressure [mPAP] >35 mmHg or mPAP 25-35 mmHg with low cardiac index [<2.0 L⋅min-1⋅m-2]). All patients received a single 20 µg dose of iloprost via a nebulizer (4.4 µg delivered at the mouthpiece). No serious adverse events were reported. Hemodynamic and gas exchange parameters (arterial blood gas and shunt fraction [Qs/Qt]) were measured or calculated at baseline and 10 min after iloprost inhalation. mPAP decreased by 2.1 mmHg (95% CI, -3.3 to -1.0), pulmonary vascular resistance (PVR) decreased by 62.4 dyn⋅s⋅cm-5 (95% CI, -92.9 to -31.8), and cardiac output increased by 0.4 L⋅min-1 (95% CI, 0.2-0.5). There was a more significant decline in PVR in patients with severe COPD-PH than in those with nonsevere COPD-PH. Hypoxemia and intrapulmonary shunt were more extreme in patients with severe COPD-PH. However, there were no significant differences in arterial blood gas and Qs/Qt between patients with nonsevere and severe forms of COPD-PH. In conclusion, iloprost improved pulmonary hemodynamics without detrimental effects on arterial oxygenation in patients with COPD-PH, even in those with severe PH. These findings suggest that the short-term use of iloprost in patients with COPD-PH is effective and well tolerated.
Collapse
Affiliation(s)
- Lan Wang
- Department of Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuan-Zhe Jin
- Department of Cardiology, The Fourth Affiliated Hospital of China Medical University, Liaoning, China
| | - Qin-Hua Zhao
- Department of Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rong Jiang
- Department of Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen-Hui Wu
- Department of Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Su-Gang Gong
- Department of Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jing He
- Department of Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jin-Ming Liu
- Department of Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhi-Cheng Jing
- Department of Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.,Thrombosis and Vascular Medicine Center, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
31
|
Wang P, Han X, Mo B, Huang G, Wang C. LPS enhances TLR4 expression and IFN‑γ production via the TLR4/IRAK/NF‑κB signaling pathway in rat pulmonary arterial smooth muscle cells. Mol Med Rep 2017; 16:3111-3116. [PMID: 28714001 PMCID: PMC5547977 DOI: 10.3892/mmr.2017.6983] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 05/16/2017] [Indexed: 01/28/2023] Open
Abstract
The aim of the present study was to investigate the role of the Toll-like receptor (TLR)4 signaling pathway in cellular response to lipopolysaccharide (LPS) in rat pulmonary artery smooth muscle cells (PASMCs). Chronic obstructive pulmonary disease (COPD) rats were established with passive inhaling cigarette smoke plus injection of LPS. The TLR4 protein in lung tissues was determined with immunohistochemical staining and protein levels of the components of the TLR4 pathway in PASMCs were analyzed with western blotting. The production of interferon (IFN)-γ upon LPS stimulation in PASMCs was measured with ELISA. TLR4 expression in lung tissue from COPD rats was increased obviously compared with that in normal group. LPS enhances TLR4 expression in rat PASMCs and induced production of IFN-γ dramatically. LPS treatment resulted in increased phosphor-interleukin-1 receptor-associated kinase (IRAK), IκB and IκB kinase, as well as the total protein of nuclear factor (NF)-κB p65. TLR4 inhibitor TAK-242, IRAK1/4 inhibitor and NF-κB inhibitor Bay 117082 were capable of suppressing the effects of LPS. TLR4 signaling pathway is functional in PASMCs, and may be involved in the inflammatory response during the pathogenesis of COPD.
Collapse
Affiliation(s)
- Pengyan Wang
- Division of Respiratory Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Xuhui Han
- Division of Respiratory Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Biwen Mo
- Division of Respiratory Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Guojin Huang
- Division of Respiratory Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Changming Wang
- Division of Respiratory Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| |
Collapse
|
32
|
Borgas D, Chambers E, Newton J, Ko J, Rivera S, Rounds S, Lu Q. Cigarette Smoke Disrupted Lung Endothelial Barrier Integrity and Increased Susceptibility to Acute Lung Injury via Histone Deacetylase 6. Am J Respir Cell Mol Biol 2017; 54:683-96. [PMID: 26452072 DOI: 10.1165/rcmb.2015-0149oc] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Epidemiologic evidence indicates that cigarette smoke (CS) is associated with the development of acute lung injury (ALI). We have previously shown that brief CS exposure exacerbates lipopolysaccharide (LPS)-induced ALI in vivo and endothelial barrier dysfunction in vitro. In this study, we found that CS also exacerbated Pseudomonas-induced ALI in mice. We demonstrated that lung microvascular endothelial cells (ECs) isolated from mice exposed to CS had a greater permeability or incomplete recovery after challenges by LPS and thrombin. Histone deacetylase (HDAC) 6 deacetylates proteins essential for maintenance of endothelial barrier function. We found that HDAC6 phosphorylation at serine-22 was increased in lung tissues of mice exposed to CS and in lung ECs exposed to cigarette smoke extract (CSE). Inhibition of HDAC6 attenuated CSE-induced increases in EC permeability and CS priming of ALI. Similar barrier protection was provided by the microtubule stabilizer taxol, which preserved α-tubulin acetylation. CSE decreased α-tubulin acetylation and caused microtubule depolymerization. In coordination with increased HDAC6 phosphorylation, CSE inhibited Akt and activated glycogen synthase kinase (GSK)-3β; these effects were ameliorated by the antioxidant N-acetyl cysteine. Our results suggest that CS increases lung EC permeability, thereby enhancing susceptibility to ALI, likely through oxidative stress-induced Akt inactivation and subsequent GSK-3β activation. Activated GSK-3β may activate HDAC6 via phosphorylation of serine-22, leading to α-tubulin deacetylation and microtubule disassembly. Inhibition of HDAC6 may be a novel therapeutic option for ALI in cigarette smokers.
Collapse
Affiliation(s)
- Diana Borgas
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Eboni Chambers
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Julie Newton
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Junsuk Ko
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Stephanie Rivera
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Sharon Rounds
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Qing Lu
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
33
|
Arian A, Moghadam SGM, Kazemi T, Hajihosseini M. The Effects of Statins on Pulmonary Artery Pressure in Patients with Chronic Obstructive Pulmonary Disease: A Randomized Controlled Trial. J Res Pharm Pract 2017; 6:27-30. [PMID: 28331863 PMCID: PMC5348853 DOI: 10.4103/2279-042x.200985] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Pulmonary hypertension is a serious complication in patients suffering from chronic obstructive pulmonary disease (COPD). The aim of this study is to investigate the effects of atorvastatin in reducing pulmonary arterial pressure in COPD patients. METHODS This double-blind, randomized trial was conducted on 42 known cases of COPD with systolic pulmonary arterial pressure of more than 25 mmHg. The patients were randomly assigned into two groups, 21 patients with atorvastatin treatment (40 mg/daily for 6 months) and 21 patients without receiving atorvastatin. All the patients participated voluntarily and provided written informed consent. The trial was registered in the Iranian Registry of Clinical Trials. Pulmonary pressure was also anticipated by Doppler echocardiography with peak pressure gradient of tricuspid regurgitation. Both groups were tested with echocardiography to measure systolic pulmonary pressure at baseline and posttreatment. Statistical analysis includes Chi-square, Student's t-test, and Wilcoxon test. P < 0.05 was considered statistically significant. FINDINGS The mean age was 65.8 ± 11.5 years for atorvastatin group and 63.7 ± 7.6 years for control group (P = 0.45). Baseline and posttreatment mean systolic pulmonary artery pressure (PAP) levels in the atorvastatin group were 48.9 ± 3.3 and 38.4 ± 1.9 mmHg, respectively (P = 0.007). In the control group, mean systolic PAP levels at baseline and 6 months later were 45.6 ± 3.1 and 38.9 ± 2.4 mmHg, respectively (P = 0.073). The patients treated with atorvastatin showed significant decrease in total cholesterol (P = 0.001) and low-density lipoprotein cholesterol (P = 0.008). CONCLUSION A 40 mg dose of atorvastatin daily for 6 months may have beneficial effects in reducing PAP in patients with COPD. Further studies are necessary to find long-term effects of statins in COPD patients.
Collapse
Affiliation(s)
- Anahita Arian
- Birjand Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Sayyed Gholamreza Mortazavi Moghadam
- Department of Internal Medicine, Pulmonary Division, Atherosclerosis and Coronary Artery Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Toba Kazemi
- Birjand Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Morteza Hajihosseini
- Birjand Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
34
|
Sutliff RL. New insights into mechanisms regulating pulmonary adventitial fibroblast proliferation. Acta Physiol (Oxf) 2017; 219:17-19. [PMID: 27420025 DOI: 10.1111/apha.12753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Roy L. Sutliff
- Department of Medicine; Emory University/Atlanta VAMC; Decatur GA USA
| |
Collapse
|
35
|
State of the Art Review of the Right Ventricle in COPD Patients: It is Time to Look Closer. Lung 2016; 195:9-17. [DOI: 10.1007/s00408-016-9961-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 11/09/2016] [Indexed: 10/20/2022]
|
36
|
Ali ER. Role of mean platelet volume in patients with chronic obstructive pulmonary disease. THE EGYPTIAN JOURNAL OF BRONCHOLOGY 2016. [DOI: 10.4103/1687-8426.193635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
37
|
Guo L, Chughtai AR, Jiang H, Gao L, Yang Y, Yang Y, Liu Y, Xie Z, Li W. Relationship between polycythemia and in-hospital mortality in chronic obstructive pulmonary disease patients with low-risk pulmonary embolism. J Thorac Dis 2016; 8:3119-3131. [PMID: 28066591 DOI: 10.21037/jtd.2016.11.31] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUNDS Pulmonary embolism (PE) is frequent in subjects with chronic obstructive pulmonary disease (COPD) and associated with high mortality. This multi-center retrospective study was performed to investigate if secondary polycythemia is associated with in-hospital mortality in COPD patients with low-risk PE. METHODS We identified COPD patients with proven PE between October, 2005 and October, 2015. Patients in risk classes III-V on the basis of the PESI score were excluded. We extracted demographic, clinical and laboratory information at the time of admission from medical records. All subjects were followed until hospital discharge to identify all-cause mortality. RESULTS We enrolled 629 consecutive patients with COPD and PE at low risk: 132 of them (21.0%) with and 497 (79.0%) without secondary polycythemia. Compared with those without polycythemia, the polycythemia group had significantly lower forced expiratory volume in one second (FEV1) level (0.9±0.3 vs. 1.4±0.5, P=0.000), lower PaO2 and SpO2 as well as higher PaCO2 (P=0.03, P=0.03 and P=0.000, respectively). COPD patients with polycythemia had a higher proportion of arrhythmia in electrocardiogram (ECG) (49.5% vs. 35.7%, P=0.02), a longer hospital duration time (15.3±10.1 vs. 9.7±9.1, P=0.001), a higher mechanical ventilation rate (noninvasive and invasive, 51.7% vs. 30.3%, P=0.04 and 31.0% vs. 7.9%, P=0.04, respectively), and a higher in-hospital mortality (12.1% vs. 6.6%, P=0.04). Multivariate logistic regression analysis revealed that polycythemia was associated with mortality in COPD patients with low-risk PE (adjusted OR 1.11; 95% CI, 1.04-1.66). CONCLUSIONS Polycythemia is an independent risk factor for all-cause in-hospital mortality in COPD patients with PE at low risk.
Collapse
Affiliation(s)
- Lu Guo
- Department of Respiratory Medicine, West China Hospital of Sichuan University, Chengdu 610041, China;; Department of Respiratory Medicine, Sichuan Academy of Medical Science, Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology of China, Chengdu 610072, China
| | | | - Hongli Jiang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Lingyun Gao
- Department of Respiratory Medicine, Sichuan Academy of Medical Science, Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology of China, Chengdu 610072, China
| | - Yan Yang
- Department of Respiratory Medicine, Sichuan Academy of Medical Science, Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology of China, Chengdu 610072, China
| | - Yang Yang
- Department of Respiratory Medicine, Sichuan Academy of Medical Science, Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology of China, Chengdu 610072, China
| | - Yuejian Liu
- Department of Respiratory Medicine, Sichuan Academy of Medical Science, Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology of China, Chengdu 610072, China
| | - Zhenliang Xie
- Department of Respiratory Medicine, Sichuan Academy of Medical Science, Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology of China, Chengdu 610072, China
| | - Weimin Li
- Department of Respiratory Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
38
|
Ju CR, Zhang JH, Chen M, Chen RC. Plasma myostatin levels are related to the extent of right ventricular dysfunction in exacerbation of chronic obstructive pulmonary disease. Biomarkers 2016; 22:246-252. [PMID: 27323660 DOI: 10.1080/1354750x.2016.1203999] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate the relationship between plasma myostatin levels and right ventricle (RV) dysfunction (RVD) in acute exacerbation of chronic obstructive pulmonary disease (AECOPD). METHODS The study recruited 84 patients with AECOPD. Plasma myostatin was analyzed and tricuspid annular plane systolic excursion (TAPSE) < 16 mm was used as the main indicator for RVD. RESULTS Plasma myostatin levels were significantly higher in 47 patients with RVD than 37 ones without (P < 0.005). Multivariate regression analysis revealed that myostatin levels correlated significantly with TAPSE values and RV myocardial performance index (p < 0.001) among the study patients. CONCLUSION Plasma myostatin is a potential biomarker for improving diagnosis of RVD in AECOPD.
Collapse
Affiliation(s)
- Chun-Rong Ju
- a State Key Lab of the Respiratory Disease, Guangzhou Institute of Respiratory Disease , First Affiliated Hospital of Guangzhou Medical University , Guangdong , China
| | - Jian-Heng Zhang
- a State Key Lab of the Respiratory Disease, Guangzhou Institute of Respiratory Disease , First Affiliated Hospital of Guangzhou Medical University , Guangdong , China
| | - Miao Chen
- a State Key Lab of the Respiratory Disease, Guangzhou Institute of Respiratory Disease , First Affiliated Hospital of Guangzhou Medical University , Guangdong , China
| | - Rong-Chang Chen
- a State Key Lab of the Respiratory Disease, Guangzhou Institute of Respiratory Disease , First Affiliated Hospital of Guangzhou Medical University , Guangdong , China
| |
Collapse
|
39
|
Caramori G, Casolari P, Barczyk A, Durham AL, Di Stefano A, Adcock I. COPD immunopathology. Semin Immunopathol 2016; 38:497-515. [PMID: 27178410 PMCID: PMC4897000 DOI: 10.1007/s00281-016-0561-5] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 04/20/2016] [Indexed: 02/06/2023]
Abstract
The immunopathology of chronic obstructive pulmonary disease (COPD) is based on the innate and adaptive inflammatory immune responses to the chronic inhalation of cigarette smoking. In the last quarter of the century, the analysis of specimens obtained from the lower airways of COPD patients compared with those from a control group of age-matched smokers with normal lung function has provided novel insights on the potential pathogenetic role of the different cells of the innate and acquired immune responses and their pro/anti-inflammatory mediators and intracellular signalling pathways, contributing to a better knowledge of the immunopathology of COPD both during its stable phase and during its exacerbations. This also has provided a scientific rationale for new drugs discovery and targeting to the lower airways. This review summarises and discusses the immunopathology of COPD patients, of different severity, compared with control smokers with normal lung function.
Collapse
Affiliation(s)
- Gaetano Caramori
- Centro Interdipartimentale per lo Studio delle Malattie Infiammatorie delle Vie Aeree e Patologie Fumo-correlate (CEMICEF; formerly named Centro di Ricerca su Asma e BPCO), Sezione di Medicina Interna e Cardiorespiratoria, Università di Ferrara, Via Savonarola 9, 44121, Ferrara, Italy.
| | - Paolo Casolari
- Centro Interdipartimentale per lo Studio delle Malattie Infiammatorie delle Vie Aeree e Patologie Fumo-correlate (CEMICEF; formerly named Centro di Ricerca su Asma e BPCO), Sezione di Medicina Interna e Cardiorespiratoria, Università di Ferrara, Via Savonarola 9, 44121, Ferrara, Italy
| | - Adam Barczyk
- Katedra i Klinika Pneumonologii, Slaski Uniwersytet Medyczny w Katowicach, Katowice, Poland
| | - Andrew L Durham
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Antonino Di Stefano
- Divisione di Pneumologia e Laboratorio di Citoimmunopatologia dell'Apparato Cardio Respiratorio, Salvatore Maugeri Foundation, IRCCS, Veruno, NO, Italy
| | - Ian Adcock
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
40
|
Grimminger J, Ghofrani HA, Weissmann N, Klose H, Grimminger F. COPD-associated pulmonary hypertension: clinical implications and current methods for treatment. Expert Rev Respir Med 2016; 10:755-66. [PMID: 27212458 DOI: 10.1080/17476348.2016.1190275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Chronic obstructive pulmonary disease is the fourth leading cause of death worldwide, one serious complication being pulmonary hypertension, which occurs in up to 30% of patients and increases mortality drastically. Difficulties in diagnosis and the unclear beneficial effects of PH-specific therapy have hitherto resulted in the absence of approved therapies. Consequently, PH and right heart failure in COPD are still currently treated according to symptoms and not underlying cause Areas covered: This review focuses on the current knowledge of its pathogenesis, clinical picture, diagnosis as well as methods for treatment Expert commentary: Since PH-COPD is an orphan disease with grievous consequences, and diagnosis as well as the right choice of possible treatment is crucial, referral to an expert center in cases of suspicion is necessary. Hitherto there is no officially approved treatment available even though several studies have shown notable improvement in selected individuals, making diagnostics, prognostic markers, and the search for therapeutic agents key issues of interest in this field.
Collapse
Affiliation(s)
- Jan Grimminger
- a University of Giessen and Marburg Lung Center (UGMLC), University of Giessen , Giessen , Germany.,b German Center for Lung Research (DZL) , University of Giessen , Giessen , Germany.,c Excellence Cluster Cardio-Pulmonary System (ECCPS) , University of Giessen , Giessen , Germany.,d University Medical Center Hamburg-Eppendorf (UKE), University of Hamburg , Hamburg , Germany.,e Center for Pulmonary Arterial Hypertension Hamburg (CPAHH), Martin Zeitz Center for Rare Diseases , University of Hamburg , Hamburg , Germany
| | - Hossein Ardeschir Ghofrani
- a University of Giessen and Marburg Lung Center (UGMLC), University of Giessen , Giessen , Germany.,b German Center for Lung Research (DZL) , University of Giessen , Giessen , Germany.,c Excellence Cluster Cardio-Pulmonary System (ECCPS) , University of Giessen , Giessen , Germany.,f Department of Medicine , Imperial College London , London , UK
| | - Nobert Weissmann
- a University of Giessen and Marburg Lung Center (UGMLC), University of Giessen , Giessen , Germany.,b German Center for Lung Research (DZL) , University of Giessen , Giessen , Germany.,c Excellence Cluster Cardio-Pulmonary System (ECCPS) , University of Giessen , Giessen , Germany
| | - Hans Klose
- d University Medical Center Hamburg-Eppendorf (UKE), University of Hamburg , Hamburg , Germany.,e Center for Pulmonary Arterial Hypertension Hamburg (CPAHH), Martin Zeitz Center for Rare Diseases , University of Hamburg , Hamburg , Germany.,g German Center for Lung Research (DZL) , University of Hamburg , Hamburg , Germany
| | - Friedrich Grimminger
- a University of Giessen and Marburg Lung Center (UGMLC), University of Giessen , Giessen , Germany.,b German Center for Lung Research (DZL) , University of Giessen , Giessen , Germany.,c Excellence Cluster Cardio-Pulmonary System (ECCPS) , University of Giessen , Giessen , Germany
| |
Collapse
|
41
|
Blanco I, Piccari L, Barberà JA. Pulmonary vasculature in COPD: The silent component. Respirology 2016; 21:984-94. [PMID: 27028849 DOI: 10.1111/resp.12772] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/06/2015] [Accepted: 12/20/2015] [Indexed: 01/15/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by airflow obstruction that results from an inflammatory process affecting the airways and lung parenchyma. Despite major abnormalities taking place in bronchial and alveolar structures, changes in pulmonary vessels also represent an important component of the disease. Alterations in vessel structure are highly prevalent and abnormalities in their function impair gas exchange and may result in pulmonary hypertension (PH), an important complication of the disease associated with reduced survival and worse clinical course. The prevalence of PH is high in COPD, particularly in advanced stages, although it remains of mild to moderate severity in the majority of cases. Endothelial dysfunction, with imbalance between vasodilator/vasoconstrictive mediators, is a key determinant of changes taking place in pulmonary vasculature in COPD. Cigarette smoke products may perturb endothelial cells and play a critical role in initiating vascular changes. The concurrence of inflammation, hypoxia and emphysema further contributes to vascular damage and to the development of PH. The use of drugs that target endothelium-dependent signalling pathways, currently employed in pulmonary arterial hypertension, is discouraged in COPD due to the lack of efficacy observed in randomized clinical trials and because there is compelling evidence indicating that these drugs may worsen pulmonary gas exchange. The subgroup of patients with severe PH should be ideally managed in centres with expertise in both PH and chronic lung diseases because alterations of pulmonary vasculature might resemble those observed in pulmonary arterial hypertension. Because this condition entails poor prognosis, it warrants specialist treatment.
Collapse
Affiliation(s)
- Isabel Blanco
- Department of Pulmonary Medicine, Hospital Clínic and August Pi i Sunyer Biomedical Research Institute (IDIBAPS); University of Barcelona and Biomedical Research Networking Center in Respiratory Diseases (CIBERES), Madrid, Spain
| | - Lucilla Piccari
- Department of Pulmonary Medicine, Hospital Clínic and August Pi i Sunyer Biomedical Research Institute (IDIBAPS); University of Barcelona and Biomedical Research Networking Center in Respiratory Diseases (CIBERES), Madrid, Spain
| | - Joan Albert Barberà
- Department of Pulmonary Medicine, Hospital Clínic and August Pi i Sunyer Biomedical Research Institute (IDIBAPS); University of Barcelona and Biomedical Research Networking Center in Respiratory Diseases (CIBERES), Madrid, Spain
| |
Collapse
|
42
|
Wang J, Zhang C, Zhang Z, Zheng Z, Sun D, Yang Q, Hadadi C, Li D, Xu X, Xiong M, Zhou Q, Guo M, Wang Y, Tang C, Xu G, Yang K, Zhong N, Lu W. A Functional Variant rs6435156C > T in BMPR2 is Associated With Increased Risk of Chronic Obstructive Pulmonary Disease (COPD) in Southern Chinese Population. EBioMedicine 2016; 5:167-74. [PMID: 27077124 PMCID: PMC4816816 DOI: 10.1016/j.ebiom.2016.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 02/01/2016] [Accepted: 02/03/2016] [Indexed: 01/26/2023] Open
Abstract
BACKGROUNDS Bone morphogenetic protein receptor type 2 (BMPR2) signaling is anti-inflammatory. Decreased BMPR2 expression was seen in lung tissue from chronic obstructive pulmonary disease (COPD) patients. METHODS The selected single nucleotide polymorphisms (SNPs) in BMPR2 were genotyped with polymerase chain reaction (PCR) ligase detection reaction. The effects of SNPs on gene expression were analyzed with luciferase assays. The mRNA and protein expression levels of BMPR2 in peripheral blood mononuclear cells (PBMCs) from COPD patients were determined by quantitative PCR and western blotting, respectively. FINDINGS Two SNPs, rs6435156C > T and rs1048829G > T in the 3'-untranslated region (3'UTR) of BMPR2 were selected and genotyped in COPD case and healthy control subjects from southern Chinese population. Both of them were found associated with significantly increased COPD risk (adjusted odds ratio [OR] = 1.58 with 95% confidence interval [CI] = 1.14-2.15, P = 0.0056 for rs6435156C > T; adjusted OR = 1.47 and 95% CI = 1.10-1.97, P = 0.0092 for rs1048829G > T). Older age, cigarette smoking, family history of cancer and COPD were all factors that interacted with rs6435156C > T and rs1048829G > T causing increased COPD risk. Cigarette smokers with rs6435156 (CT + TT) or rs1048829 (GT + TT) were more susceptible to COPD than that with the rs6435156CC or rs1048829GG genotypes. In A549 human alveolar epithelial cells, luciferase reporter assays revealed that introduction of 3'UTR of BMPR2 plasmids carrying rs6435156T allele but not rs1048829T led to lower luciferase activity than the wild-type C or G alleles. Comparing to rs6435156CC, treatment with hsa-miR-20a mimics deceased whereas hsa-miR-20a inhibitor restored the luciferase reporter activity in cells transfected with constructs carrying rs6435156TT. BMPR2 mRNA and protein expressions were significantly lower in PBMCs from COPD smokers than that in non-smokers. COPD patients carrying rs6435156T allele had less BMPR2 expression in PBMCs. INTERPRETATION This study demonstrated that both rs6435156C > T and rs1048829G > T variants in BMPR2 contributed to increased susceptibility to COPD. The T variants of rs6435156 increased COPD risk likely by binding with hsa-miR-20a, thus leading to downregulated BMPR2 expression in lung epithelial and immune cells.
Collapse
Affiliation(s)
- Jian Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Department of Respiration, Inner Mongolia Autonomous Region People's Hospital, Hohhot 010017, Inner Mongolia, China; Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Chenting Zhang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zili Zhang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zeguang Zheng
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dejun Sun
- Department of Respiration, Inner Mongolia Autonomous Region People's Hospital, Hohhot 010017, Inner Mongolia, China
| | - Quan Yang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cyrus Hadadi
- Geisinger Medical Center, 100 North Academy Avenue, Danville, PA 17822, USA
| | - Defu Li
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoming Xu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingmei Xiong
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qipeng Zhou
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Meihua Guo
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yingfeng Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chun Tang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guihua Xu
- Department of Respiration, Inner Mongolia Autonomous Region People's Hospital, Hohhot 010017, Inner Mongolia, China
| | - Kai Yang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
43
|
Terada M, Takehara Y, Isoda H, Uto T, Matsunaga M, Alley M. Low WSS and High OSI Measured by 3D Cine PC MRI Reflect High Pulmonary Artery Pressures in Suspected Secondary Pulmonary Arterial Hypertension. Magn Reson Med Sci 2015; 15:193-202. [PMID: 26567758 PMCID: PMC5600056 DOI: 10.2463/mrms.mp.2015-0038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Purpose: Institutional Review Board (IRB)-approved prospective study was conducted to test whether objective and quantitative hemodynamic markers wall shear stress (WSS) and oscillatory shear index (OSI) measured by three-dimensional (3D) cine phase-contrast (PC) can reflect pulmonary arterial hypertension (PAH). Patients and Methods: Seventeen consecutive patients of suspected secondary PAH were examined for pulmonary artery pressures (PAPs) with right heart catheterization (RHC) and three-dimensional (3D) cine PC MR. Based on the RHC data, patients were subdivided into two groups of 12 non-PAH (median age of 74.5 years) and 5 PAH (median age of 77 years) patients. Based on 3D cine PC magnetic resonance (MR), hemodynamic parameters including spatially averaged systolic WSS (sWSS), diastolic WSS (dWSS), mean WSS (mWSS), OSI and blood vessel section area (BVSA) at the pulmonary arterial trunk were calculated. Streamline images in the pulmonary arteries were also assessed. All the parameters were compared between non-PAH and PAH groups. Results: sWSS (N/m2) and mWSS (N/m2) of PAH group was lower than that of non-PAH group (0.594 ± 0.067 vs. 0.961 ± 0.590, P = 0.001), (0.365 ± 0.035 vs. 0.489 ± 0.132, P = 0.027). OSI of PAH group was higher than that of non-PAH (0.214 ± 0.026 vs. 0.130 ± 0.046, P = 0.001). sWSS, mWSS, and dWSS were inversely correlated and OSI was positively correlated to mean PAP or systolic PAP with r values of –0.638 (P = 0.005), –0.643 (P = 0.005), –0.485 (P = 0.049), and 0.625 (P = 0.007); or –0.622 (P = 0.008), –0.629 (P = 0.007), –0.484 (P = 0.049), and 0.594 (P = 0.012), respectively. sWSS was also inversely correlated to BVSA with r value of –0.488 (P = 0.049), and OSI was correlated to BVSA with r value of 0.574 (P = 0.016). Vortex or helical flows were observed more frequently in PAH patients. Conclusions: The low sWSS and mWSS as well as high OSI measured with 3D cine PC MR could be potential hemodynamic markers for the increased PAP in suspected secondary PAH patients.
Collapse
Affiliation(s)
- Masaki Terada
- Department of Diagnostic Radiological Technology, Iwata City Hospital
| | | | | | | | | | | |
Collapse
|
44
|
Detection of right sided heart changes and pulmonary hypertension in COPD patients. EGYPTIAN JOURNAL OF CHEST DISEASES AND TUBERCULOSIS 2015. [DOI: 10.1016/j.ejcdt.2014.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
45
|
Robinson JC, Graham BB, Rouault TC, Tuder RM. The crossroads of iron with hypoxia and cellular metabolism. Implications in the pathobiology of pulmonary hypertension. Am J Respir Cell Mol Biol 2015; 51:721-9. [PMID: 24988529 DOI: 10.1165/rcmb.2014-0021tr] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The pathologic hallmark of pulmonary arterial hypertension (PAH) is pulmonary vascular remodeling, characterized by endothelial cell proliferation, smooth muscle hypertrophy, and perivascular inflammation, ultimately contributing to increased pulmonary arterial pressures. Several recent studies have observed that iron deficiency in patients with various forms of PAH is associated with worsened clinical outcome. Iron plays a key role in many cellular processes regulating the response to hypoxia, oxidative stress, cellular proliferation, and cell metabolism. Given the potential importance of iron supplementation in patients with the disease and the broad cellular functions of iron, we review its role in processes that pertain to PAH.
Collapse
Affiliation(s)
- Jeffrey C Robinson
- 1 Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado; and
| | | | | | | |
Collapse
|
46
|
Weissmann N, Lobo B, Pichl A, Parajuli N, Seimetz M, Puig-Pey R, Ferrer E, Peinado VI, Domínguez-Fandos D, Fysikopoulos A, Stasch JP, Ghofrani HA, Coll-Bonfill N, Frey R, Schermuly RT, García-Lucio J, Blanco I, Bednorz M, Tura-Ceide O, Tadele E, Brandes RP, Grimminger J, Klepetko W, Jaksch P, Rodriguez-Roisin R, Seeger W, Grimminger F, Barberà JA. Stimulation of soluble guanylate cyclase prevents cigarette smoke-induced pulmonary hypertension and emphysema. Am J Respir Crit Care Med 2014; 189:1359-73. [PMID: 24738736 DOI: 10.1164/rccm.201311-2037oc] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
RATIONALE Chronic obstructive pulmonary disease (COPD) is a major cause of death worldwide. No therapy stopping progress of the disease is available. OBJECTIVES To investigate the role of the soluble guanylate cyclase (sGC)-cGMP axis in development of lung emphysema and pulmonary hypertension (PH) and to test whether the sGC-cGMP axis is a treatment target for these conditions. METHODS Investigations were performed in human lung tissue from patients with COPD, healthy donors, mice, and guinea pigs. Mice were exposed to cigarette smoke (CS) for 6 hours per day, 5 days per week for up to 6 months and treated with BAY 63-2521. Guinea pigs were exposed to CS from six cigarettes per day for 3 months, 5 days per week and treated with BAY 41-2272. Both BAY compounds are sGC stimulators. Gene and protein expression analysis were performed by quantitative real-time polymerase chain reaction and Western blotting. Lung compliance, hemodynamics, right ventricular heart mass alterations, and alveolar and vascular morphometry were performed, as well as inflammatory cell infiltrate assessment. In vitro assays of cell adhesion, proliferation, and apoptosis have been done. MEASUREMENTS AND MAIN RESULTS The functionally essential sGC β1-subunit was down-regulated in patients with COPD and in CS-exposed mice. sGC stimulators prevented the development of PH and emphysema in the two different CS-exposed animal models. sGC stimulation prevented peroxynitrite-induced apoptosis of alveolar and endothelial cells, reduced CS-induced inflammatory cell infiltrate in lung parenchyma, and inhibited adhesion of CS-stimulated neutrophils. CONCLUSIONS The sGC-cGMP axis is perturbed by chronic exposure to CS. Treatment of COPD animal models with sGC stimulators can prevent CS-induced PH and emphysema.
Collapse
Affiliation(s)
- Norbert Weissmann
- 1 Justus-Liebig University, Excellence Cluster Cardiopulmonary System, Universities of Giessen and Marburg Lung Center (UGMLC), DZL, Giessen, Marburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
[Pulmonary hypertension in chronic respiratory diseases]. Presse Med 2014; 43:945-56. [PMID: 25123317 DOI: 10.1016/j.lpm.2014.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 07/09/2014] [Indexed: 11/23/2022] Open
Abstract
Pulmonary hypertension is frequent in advanced chronic respiratory diseases, with an estimated prevalence at the time of pulmonary transplantation of 30-50 % in idiopathic pulmonary fibrosis, 30-50 % in chronic obstructive pulmonary disease, 50 % in combined pulmonary fibrosis and emphysema, 75 % in sarcoidosis, and more than 75 % of cases in pulmonary Langerhans cell histiocytosis. Histologic features include varying degrees of pulmonary arterial remodeling (prominent), vascular rarefaction (emphysema), fibrosis or specific involvement of the pulmonary arteries (idiopathic pulmonary fibrosis, sarcoidosis, lymphangioleiomyomatosis, pulmonary Langerhans cell histiocytosis), in situ thrombosis, and frequently associated involvement of the pulmonary veins (idiopathic pulmonary fibrosis, sarcoidosis). Pulmonary hypertension is usually detected using echocardiography with Doppler, however right heart catheterisation is required to confirm precapillary pulmonary hypertension defined by pulmonary artery pressure ≥ 25 mm Hg, with pulmonary artery wedge pressure ≤ 15 mm Hg. When present, it is associated with decreased exercise capacity and worse mortality. Pulmonary hypertension in chronic respiratory disease is almost invariably multifactorial; hypoxia is one of its main determinants, however supplemental oxygen therapy rarely reverses pulmonary hypertension. Management of pulmonary hypertension in chronic respiratory disease is mostly based on the optimal treatment of the underlying disease. Available data do not support the use of drug therapies specific for pulmonary hypertension in the setting of chronic respiratory diseases, however very few clinical studies have been conducted so far specifically in this context.
Collapse
|
48
|
Park YM, Chung WJ, Lee SP, Choi DY, Baek HJ, Jung SH, Choi IS, Shin EK. Efficacy of inhaled iloprost in cor pulmonale and severe pulmonary hypertension associated with tuberculous destroyed lung. J Cardiovasc Ultrasound 2014; 22:95-7. [PMID: 25031802 PMCID: PMC4096673 DOI: 10.4250/jcu.2014.22.2.95] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Revised: 04/02/2014] [Accepted: 05/15/2014] [Indexed: 11/22/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the causes of cor pulmonale. Cor pulmonale patients with pulmonary hypertension have a significant lower survival rate than patients without. However, there is no conclusive treatment options in cor pulmonale and pulmonary hypertension associated with COPD until now. We report a patient with cor pulmonale and pulmonary hypertension associated with severe form of COPD and tuberculous destroyed lung who achieved marked clinical, functional and echocardiographic hemodynamic improvements with inhaled iloprost for six months.
Collapse
Affiliation(s)
- Yae Min Park
- Division of Cardiology, Heart Center, Gachon University Gil Medical Center, Incheon, Korea. ; Gachon Cardiovascular Research Institute, Gachon University Gil Medical Center, Incheon, Korea
| | - Wook-Jin Chung
- Division of Cardiology, Heart Center, Gachon University Gil Medical Center, Incheon, Korea. ; Gachon Cardiovascular Research Institute, Gachon University Gil Medical Center, Incheon, Korea
| | - Sang Pyo Lee
- Division of Pulmonology, Gachon University Gil Medical Center, Incheon, Korea
| | - Deok Young Choi
- Gachon Cardiovascular Research Institute, Gachon University Gil Medical Center, Incheon, Korea. ; Department of Pediatric Cardiology, Gachon University Gil Medical Center, Incheon, Korea
| | - Han Joo Baek
- Division of Rheumatology, Gachon University Gil Medical Center, Incheon, Korea
| | - Sung Hwan Jung
- Division of Pulmonology, Gachon University Gil Medical Center, Incheon, Korea
| | - In Suck Choi
- Division of Cardiology, Heart Center, Gachon University Gil Medical Center, Incheon, Korea. ; Gachon Cardiovascular Research Institute, Gachon University Gil Medical Center, Incheon, Korea
| | - Eak Kyun Shin
- Division of Cardiology, Heart Center, Gachon University Gil Medical Center, Incheon, Korea. ; Gachon Cardiovascular Research Institute, Gachon University Gil Medical Center, Incheon, Korea
| |
Collapse
|
49
|
Iyer AS, Wells JM, Vishin S, Bhatt SP, Wille KM, Dransfield MT. CT scan-measured pulmonary artery to aorta ratio and echocardiography for detecting pulmonary hypertension in severe COPD. Chest 2014; 145:824-832. [PMID: 24114440 DOI: 10.1378/chest.13-1422] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND COPD is associated with significant morbidity primarily driven by acute exacerbations. Relative pulmonary artery (PA) enlargement, defined as a PA to ascending aorta (A) diameter ratio greater than one (PA:A>1) identifies patients at increased risk for exacerbations. However, little is known about the correlation between PA:A, echocardiography, and invasive hemodynamics in COPD. METHODS A retrospective observational study of patients with severe COPD being evaluated for lung transplantation at a single center between 2007 and 2011 was conducted. Clinical characteristics, CT scans, echocardiograms, and right-sided heart catheterizations were reviewed. The PA diameter at the bifurcation and A diameter from the same CT image were measured. Linear and logistic regression were used to examine the relationships between PA:A ratio by CT scan and PA systolic pressure (PASP) by echocardiogram with invasive hemodynamics. Receiver operating characteristic analysis assessed the usefulness of the PA:A ratio and PASP in predicting resting pulmonary hypertension (PH) (mean pulmonary artery pressure [mPAP]>25 mm Hg). RESULTS Sixty patients with a mean predicted FEV1 of 27%±12% were evaluated. CT scan-measured PA:A correlated linearly with mPAP after adjustment for multiple covariates (r=0.30, P=.03), a finding not observed with PASP. In a multivariate logistic model, mPAP was independently associated with PA:A>1 (OR, 1.44; 95% CI, 1.02-2.04; P=.04). PA:A>1 was 73% sensitive and 84% specific for identifying patients with resting PH (area under the curve, 0.83; 95% CI, 0.72-0.93; P<.001), whereas PASP was not useful. CONCLUSIONS A PA:A ratio>1 on CT scan outperforms echocardiography for diagnosing resting PH in patients with severe COPD.
Collapse
Affiliation(s)
- Anand S Iyer
- Department of Internal Medicine, University of Alabama at Birmingham
| | - J Michael Wells
- Department of Internal Medicine, University of Alabama at Birmingham; Division of Pulmonary, Allergy, and Critical Care, Department of Medicine and University of Alabama at Birmingham Lung Health Center, University of Alabama at Birmingham; Birmingham Veterans Affairs Medical Center, Birmingham, AL.
| | - Sonia Vishin
- Department of Internal Medicine, University of Alabama at Birmingham; Division of Pulmonary, Allergy, and Critical Care, Department of Medicine and University of Alabama at Birmingham Lung Health Center, University of Alabama at Birmingham
| | - Surya P Bhatt
- Department of Internal Medicine, University of Alabama at Birmingham
| | - Keith M Wille
- Department of Internal Medicine, University of Alabama at Birmingham; Division of Pulmonary, Allergy, and Critical Care, Department of Medicine and University of Alabama at Birmingham Lung Health Center, University of Alabama at Birmingham
| | - Mark T Dransfield
- Department of Internal Medicine, University of Alabama at Birmingham; Division of Pulmonary, Allergy, and Critical Care, Department of Medicine and University of Alabama at Birmingham Lung Health Center, University of Alabama at Birmingham; Birmingham Veterans Affairs Medical Center, Birmingham, AL
| |
Collapse
|
50
|
Abstract
Chronic obstructive lung disease (COPD) and diffuse parenchymal lung diseases (DPLD), including idiopathic pulmonary fibrosis (IPF) and sarcoidosis, are associated with a high incidence of pulmonary hypertension (PH), which is linked with exercise limitation and a worse prognosis. Patients with combined pulmonary fibrosis and emphysema (CPFE) are particularly prone to the development of PH. Echocardiography and right heart catheterization are the principal modalities for the diagnosis of COPD and DPLD. For discrimination between group 1 PH patients with concomitant respiratory abnormalities and group 3 PH patients (PH caused by lung disease), patients should be transferred to a center with expertise in both PH and lung diseases for comprehensive evaluation. The task force encompassing the authors of this article provided criteria for this discrimination and suggested using the following definitions for group 3 patients, as exemplified for COPD, IPF, and CPFE: COPD/IPF/CPFE without PH (mean pulmonary artery pressure [mPAP] <25 mm Hg); COPD/IPF/CPFE with PH (mPAP ≥25 mm Hg); PH-COPD, PH-IPF, and PH-CPFE); COPD/IPF/CPFE with severe PH (mPAP ≥35 mm Hg or mPAP ≥25 mm Hg with low cardiac index [CI <2.0 l/min/m(2)]; severe PH-COPD, severe PH-IPF, and severe PH-CPFE). The "severe PH group" includes only a minority of chronic lung disease patients who are suspected of having strong general vascular abnormalities (remodeling) accompanying the parenchymal disease and with evidence of an exhausted circulatory reserve rather than an exhausted ventilatory reserve underlying the limitation of exercise capacity. Exertional dyspnea disproportionate to pulmonary function tests, low carbon monoxide diffusion capacity, and rapid decline of arterial oxygenation upon exercise are typical clinical features of this subgroup with poor prognosis. Studies evaluating the effect of pulmonary arterial hypertension drugs currently not approved for group 3 PH patients should focus on this severe PH group, and for the time being, these patients should be transferred to expert centers for individualized patient care.
Collapse
|