1
|
Adachi T, Adachi S, Nakano Y, Nishiyama I, Hirose M, Murohara T. Controlling Nutritional Status Score Predicts 1-Year Outcomes in Chronic Thromboembolic Pulmonary Hypertension. Circ Rep 2024; 6:381-388. [PMID: 39262638 PMCID: PMC11383544 DOI: 10.1253/circrep.cr-24-0023] [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] [Received: 03/13/2024] [Revised: 06/27/2024] [Accepted: 07/13/2024] [Indexed: 09/13/2024] Open
Abstract
Background The prognosis for patients with chronic thromboembolic pulmonary hypertension (CTEPH) using their nutritional status has not been established. We investigated the relationship between the prognosis of patients with CTEPH and the Controlling Nutritional Status (CONUT) score, which is a nutritional assessment tool. Methods and Results A total of 157 patients with CTEPH was enrolled in the study. The primary outcome was defined as the composite outcome of all-cause mortality and non-elective hospitalization due to heart failure. Receiver operating characteristic (ROC) curve analysis was used to determine the cutoff CONUT score for predicting the 1-year rate of the primary outcome. Patients were divided into 2 groups according to the significant cutoff value and compared. Undernutrition was observed in 51.6% of patients. ROC analysis revealed a significant cutoff CONUT score of 3.5 (area under the curve=0.789). The incidence rate of the primary composite outcome was higher in the high CONUT group (score ≥4) than in the low CONUT group (score ≤3; 20% vs. 2.2%; P<0.001). Cox analysis revealed the CONUT score per point increase was an independent risk factor for the primary composite outcomes (hazard ratio 2.301; 95% confidence interval 1.081-4.895; P=0.031). Conclusions The CONUT score can predict the 1-year rate of all-cause death and non-elective hospitalization in patients with CTEPH.
Collapse
Affiliation(s)
- Takeshi Adachi
- Department of Cardiology, Nagoya University Hospital Nagoya Japan
| | - Shiro Adachi
- Department of Cardiology, Nagoya University Hospital Nagoya Japan
| | - Yoshihisa Nakano
- Center for Advanced Medicine and Clinical Research, Department of Advanced Medicine, Nagoya University Hospital Nagoya Japan
| | - Itsumure Nishiyama
- Department of Cardiology, Nagoya University Graduate School of Medicine Nagoya Japan
| | - Miku Hirose
- Department of Cardiology, Nagoya University Graduate School of Medicine Nagoya Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Hospital Nagoya Japan
| |
Collapse
|
2
|
Namazi M, Eftekhar SP, Mosaed R, Shiralizadeh Dini S, Hazrati E. Pulmonary Hypertension and Right Ventricle: A Pathophysiological Insight. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2024; 18:11795468241274744. [PMID: 39257563 PMCID: PMC11384539 DOI: 10.1177/11795468241274744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 07/21/2024] [Indexed: 09/12/2024]
Abstract
Background Pulmonary hypertension (PH) is a pulmonary vascular disease characterized by elevated pulmonary vascular pressure. Long-term PH, irrespective of its etiology, leads to increased right ventricular (RV) pressure, RV hypertrophy, and ultimately, RV failure. Main body Research indicates that RV failure secondary to hypertrophy remains the primary cause of mortality in pulmonary arterial hypertension (PAH). However, the impact of PH on RV structure and function under increased overload remains incompletely understood. Several mechanisms have been proposed, including extracellular remodeling, RV hypertrophy, metabolic disturbances, inflammation, apoptosis, autophagy, endothelial-to-mesenchymal transition, neurohormonal dysregulation, capillary rarefaction, and ischemia. Conclusions Studies have demonstrated the significant role of oxidative stress in the development of RV failure. Understanding the interplay among these mechanisms is crucial for the prevention and management of RV failure in patients with PH.
Collapse
Affiliation(s)
- Mehrshad Namazi
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
- Clinical Biomechanics and Ergonomics Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Seyed Parsa Eftekhar
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Mosaed
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| | | | - Ebrahim Hazrati
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Mason T, Mukherjee B, Marino P. Pulmonary Hypertension and the Gut Microbiome. Biomedicines 2024; 12:169. [PMID: 38255274 PMCID: PMC10813515 DOI: 10.3390/biomedicines12010169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
The gut microbiome and its associated metabolites are integral to the maintenance of gut integrity and function. There is increasing evidence that its alteration, referred to as dysbiosis, is involved in the development of a systemic conditions such as cardiovascular disease (e.g., systemic hypertension, atherosclerosis). Pulmonary hypertension (PH) is a condition characterised by progressive remodelling and vasoconstriction of the pulmonary circulation, ultimately leading to right ventricular failure and premature mortality if untreated. Initial studies have suggested a possible association between dysbiosis of the microbiome and the development of PH. The aim of this article is to review the current experimental and clinical data with respect to the potential interaction between the gut microbiome and the pathophysiology of pulmonary hypertension. It will also highlight possible new therapeutic targets that may provide future therapies.
Collapse
Affiliation(s)
- Thomas Mason
- Lane Fox Respiratory Service, Guy’s & St Thomas’ Hospital NHS Foundation Trust, London SE1 7EH, UK
| | - Bhashkar Mukherjee
- Lane Fox Respiratory Service, Guy’s & St Thomas’ Hospital NHS Foundation Trust, London SE1 7EH, UK
- National Pulmonary Hypertension Service, Royal Brompton Hospital, London SW3 6NP, UK
| | - Philip Marino
- Lane Fox Respiratory Service, Guy’s & St Thomas’ Hospital NHS Foundation Trust, London SE1 7EH, UK
| |
Collapse
|
4
|
Elhage Hassan M, Vinales J, Perkins S, Sandesara P, Aggarwal V, Jaber WA. Pathogenesis, Diagnosis, and Management of Chronic Thromboembolic Pulmonary Hypertension. Interv Cardiol Clin 2023; 12:e37-e49. [PMID: 38964822 DOI: 10.1016/j.iccl.2024.04.003] [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] [Indexed: 07/06/2024]
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is thought to occur as a sequelae of thromboembolic processes in the pulmonary vasculature. The pathophysiology of CTEPH is multifactorial, including impaired fibrinolysis, endothelial dysregulation, and hypoxic adaptations. The diagnosis of CTEPH is typically delayed considering the nonspecific nature of the symptoms, lack of screening, and relatively low incidence. Diagnostic tools include ventilation-perfusion testing, echocardiography, cardiac catheterization, and pulmonary angiography. The only potentially curative treatment for CTEPH is pulmonary endarterectomy However, approximately 40% of patients are inoperable. Currently, only Riociguat is Food and Drug Administration approved specifically for CTEPH, with additional drug trials underway.
Collapse
Affiliation(s)
- Malika Elhage Hassan
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road Northeast Suite F606, Atlanta, GA 30322, USA
| | - Jorge Vinales
- Department of Medicine, University of Michigan Medical School, 1301 Catherine Street, Ann Arbor, MI 48109, USA
| | - Sidney Perkins
- Department of Internal Medicine, University of Michigan Medical School, 1500 E Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Pratik Sandesara
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road Northeast Suite F606, Atlanta, GA 30322, USA
| | - Vikas Aggarwal
- Department of Cardiology, Henry Ford Medical Center, 2799 W Grand Blvd, K-2 Cath Admin Suite, Detroit, MI 48206, USA
| | - Wissam A Jaber
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road Northeast Suite F606, Atlanta, GA 30322, USA.
| |
Collapse
|
5
|
Bekedam FT, Goumans MJ, Bogaard HJ, de Man FS, Llucià-Valldeperas A. Molecular mechanisms and targets of right ventricular fibrosis in pulmonary hypertension. Pharmacol Ther 2023; 244:108389. [PMID: 36940790 DOI: 10.1016/j.pharmthera.2023.108389] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/19/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023]
Abstract
Right ventricular fibrosis is a stress response, predominantly mediated by cardiac fibroblasts. This cell population is sensitive to increased levels of pro-inflammatory cytokines, pro-fibrotic growth factors and mechanical stimulation. Activation of fibroblasts results in the induction of various molecular signaling pathways, most notably the mitogen-activated protein kinase cassettes, leading to increased synthesis and remodeling of the extracellular matrix. While fibrosis confers structural protection in response to damage induced by ischemia or (pressure and volume) overload, it simultaneously contributes to increased myocardial stiffness and right ventricular dysfunction. Here, we review state-of-the-art knowledge of the development of right ventricular fibrosis in response to pressure overload and provide an overview of all published preclinical and clinical studies in which right ventricular fibrosis was targeted to improve cardiac function.
Collapse
Affiliation(s)
- F T Bekedam
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - M J Goumans
- Department of Cell and Chemical Biology, Leiden UMC, 2300 RC Leiden, the Netherlands
| | - H J Bogaard
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - F S de Man
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands.
| | - A Llucià-Valldeperas
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands.
| |
Collapse
|
6
|
Qin X, Lei C, Yan L, Sun H, Liu X, Guo Z, Sun W, Guo X, Fang Q. Proteomic and Metabolomic Analyses of Right Ventricular Failure due to Pulmonary Arterial Hypertension. Front Mol Biosci 2022; 9:834179. [PMID: 35865003 PMCID: PMC9294162 DOI: 10.3389/fmolb.2022.834179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/20/2022] [Indexed: 11/23/2022] Open
Abstract
Right ventricular failure (RVF) is the independent and strongest predictor of mortality in pulmonary arterial hypertension (PAH), but, at present, there are no preventive and therapeutic strategies directly targeting the failing right ventricle (RV). The underlying mechanism of RV hypertrophy (RVH) and dysfunction needs to be explored in depth. In this study, we used myocardial proteomics combined with metabolomics to elucidate potential pathophysiological changes of RV remodeling in a monocrotaline (MCT)-induced PAH rat model. The proteins and metabolites extracted from the RV myocardium were identified using label-free liquid chromatography–tandem mass spectrometry (LC-MS/MS). The bioinformatic analysis indicated that elevated intracellular Ca2+ concentrations and inflammation may contribute to myocardial proliferation and contraction, which may be beneficial for maintaining the compensated state of the RV. In the RVF stage, ferroptosis, mitochondrial metabolic shift, and insulin resistance are significantly involved. Dysregulated iron homeostasis, glutathione metabolism, and lipid peroxidation related to ferroptosis may contribute to RV decompensation. In conclusion, we depicted a proteomic and metabolomic profile of the RV myocardium during the progression of MCT-induced PAH, and also provided the insights for potential therapeutic targets facilitating the retardation or reversal of RV dysfunction in PAH.
Collapse
Affiliation(s)
- Xiaohan Qin
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Chuxiang Lei
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Li Yan
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Haidan Sun
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xiaoyan Liu
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Zhengguang Guo
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wei Sun
- Core Facility of Instrument, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xiaoxiao Guo
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: Xiaoxiao Guo, ; Quan Fang,
| | - Quan Fang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: Xiaoxiao Guo, ; Quan Fang,
| |
Collapse
|
7
|
Changes in Inflammatory Markers in Patients with Chronic Thromboembolic Pulmonary Hypertension Treated with Balloon Pulmonary Angioplasty. Cells 2022; 11:cells11091491. [PMID: 35563797 PMCID: PMC9102042 DOI: 10.3390/cells11091491] [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] [Received: 01/20/2022] [Revised: 03/28/2022] [Accepted: 04/19/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Inflammatory response and endothelial dysfunction contribute to the progression of chronic thromboembolic pulmonary hypertension (CTEPH). We aimed to assess changes in biomarkers involved in those processes in inoperable CTEPH patients treated with balloon pulmonary angioplasty (BPA). Methods: We enrolled 20 patients with inoperable CTEPH qualified for BPA and a control group. Interleukin 6, 8, 10 (IL-6, IL-8, IL-10), monocyte chemoattractant protein-1 (MCP-1), and C-reactive protein (hsCRP) constituted the markers of systemic inflammation. Endothelin 1 (ET-1) served as a marker of endothelial dysfunction. Selected markers were assessed before the BPA treatment, 24 h after the first BPA, and six months after completion of the BPA treatment. Results: At baseline, the CTEPH patients had increased serum concentrations of IL-6, IL-8 and ET-1. Twenty-four hours after a BPA session, we observed an increase in concentrations of IL-6 (∆ = 3.67 (1.41; 7.16); p < 0.001), of IL-10 (∆ = 0.25 (0; 0.47); p = 0.003), of MCP-1 (∆ = 111 (60.1; 202.8); p = 0.002), and of hsCRP (∆ = 4.81 (3.46; 8.47); p < 0.001). Six months after completion of the BPA treatment, there was a decrease in concentrations of IL-6 (∆ = −1.61 (−3.11; −0.20); p = 0.03), of IL8 (∆ = −3.24 (−7.72; 0.82); p = 0.01), and of ET-1 (∆ = −0.47 (−0.96; 0.05); p = 0.005). Conclusions: Patients with inoperable CTEPH exhibit increased systemic inflammation and endothelial dysfunction, which improves after completion of the BPA treatment. A single BPA session evokes an acute inflammatory response.
Collapse
|
8
|
Ikubo Y, Sanada TJ, Hosomi K, Park J, Naito A, Shoji H, Misawa T, Suda R, Sekine A, Sugiura T, Shigeta A, Nanri H, Sakao S, Tanabe N, Mizuguchi K, Kunisawa J, Suzuki T, Tatsumi K. Altered gut microbiota and its association with inflammation in patients with chronic thromboembolic pulmonary hypertension: a single-center observational study in Japan. BMC Pulm Med 2022; 22:138. [PMID: 35395844 PMCID: PMC8994357 DOI: 10.1186/s12890-022-01932-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/01/2022] [Indexed: 11/10/2022] Open
Abstract
Background The pathogenesis of chronic thromboembolic pulmonary hypertension (CTEPH) is considered to be associated with chronic inflammation; however, the underlying mechanism remains unclear. Recently, altered gut microbiota were found in patients with pulmonary arterial hypertension (PAH) and in experimental PAH models. The aim of this study was to characterize the gut microbiota in patients with CTEPH and assess the relationship between gut dysbiosis and inflammation in CTEPH. Methods In this observational study, fecal samples were collected from 11 patients with CTEPH and 22 healthy participants. The abundance of gut microbiota in these fecal samples was assessed using 16S ribosomal ribonucleic acid (rRNA) gene sequencing. Inflammatory cytokine and endotoxin levels were also assessed in patients with CTEPH and control participants. Results The levels of serum tumor necrosis factor-α (TNF-α), interleukin (IL)-6, IL-8, and macrophage inflammatory protein (MIP)-1α were elevated in patients with CTEPH. Plasma endotoxin levels were significantly increased in patients with CTEPH (P < 0.001), and were positively correlated with TNF-α, IL-6, IL-8, and MIP-1α levels. The 16S rRNA gene sequencing and the principal coordinate analysis revealed the distinction in the gut microbiota between patients with CTEPH (P < 0.01) and control participants as well as the decreased bacterial alpha-diversity in patients with CTEPH. A random forest analysis for predicting the distinction in gut microbiota revealed an accuracy of 80.3%. Conclusion The composition of the gut microbiota in patients with CTEPH was distinct from that of healthy participants, which may be associated with the elevated inflammatory cytokines and endotoxins in CTEPH. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-022-01932-0.
Collapse
Affiliation(s)
- Yumiko Ikubo
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Takayuki Jujo Sanada
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan.
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health, and Nutrition, Osaka, Japan
| | - Jonguk Park
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Akira Naito
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Hiroki Shoji
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Tomoko Misawa
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Rika Suda
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan.,Department of Respirology, Chibaken Saiseikai Narashino Hospital, Narashino, Japan
| | - Ayumi Sekine
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Toshihiko Sugiura
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Ayako Shigeta
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Hinako Nanri
- Section of Energy Metabolism, Department of Nutrition and Metabolism, National Institute of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Nobuhiro Tanabe
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan.,Department of Respirology, Chibaken Saiseikai Narashino Hospital, Narashino, Japan
| | - Kenji Mizuguchi
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Institute for Protein Research, Osaka University, Osaka, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health, and Nutrition, Osaka, Japan
| | - Takuji Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| |
Collapse
|
9
|
Liang S, Desai AA, Black SM, Tang H. Cytokines, Chemokines, and Inflammation in Pulmonary Arterial Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:275-303. [PMID: 33788198 DOI: 10.1007/978-3-030-63046-1_15] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
According to the World Symposium Pulmonary Hypertension (WSPH) classification, pulmonary hypertension (PH) is classified into five categories based on etiology. Among them, Group 1 pulmonary arterial hypertension (PAH) disorders are rare but progressive and often, fatal despite multiple approved treatments. Elevated pulmonary arterial pressure in patients with WSPH Group 1 PAH is mainly caused by increased pulmonary vascular resistance (PVR), due primarily to sustained pulmonary vasoconstriction and excessive obliterative pulmonary vascular remodeling. Growing evidence indicates that inflammation plays a critical role in the development of pulmonary vascular remodeling associated with PAH. While the role of auto-immunity is unclear, infiltration of inflammatory cells in and around vascular lesions, including T- and B-cells, dendritic cells, macrophages, and mast cells have been observed in PAH patients. Serum and plasma levels of chemokines, cytokines, and autoantibodies are also increased in PAH patients; some of these circulating molecules are correlated with disease severity and survival. Preclinical experiments have reported a key role of the inflammation in PAH pathophysiology in vivo. Importantly, anti-inflammatory and immunosuppressive agents have further exhibited therapeutic effects. The present chapter reviews published experimental and clinical evidence highlighting the canonical role of inflammation in the pathogenesis of PAH and as a major target for the development of anti-inflammatory therapies in patients with PAH.
Collapse
Affiliation(s)
- Shuxin Liang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ankit A Desai
- Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Stephen M Black
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Haiyang Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China. .,State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
10
|
Wang Y, Huang XX, Leng D, Li JF, Liang Y, Jiang T. Effect of EZH2 on pulmonary artery smooth muscle cell migration in pulmonary hypertension. Mol Med Rep 2020; 23:129. [PMID: 33313943 PMCID: PMC7751464 DOI: 10.3892/mmr.2020.11768] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 10/27/2020] [Indexed: 12/22/2022] Open
Abstract
Pulmonary hypertension (PH) is a life‑threatening disease that often involves vascular remodeling. Although pulmonary arterial smooth muscle cells (PASMCs) are the primary participants in vascular remodeling, their biological role is not entirely clear. The present study analyzed the role of enhancer of zeste homolog 2 (EZH2) in vascular remodeling of PH by investigating the behavior of PASMCs. The expression levels of EZH2 in PASMCs in chronic thromboembolic pulmonary hypertension (CTEPH), a type of PH, were detected. The role of EZH2 in PASMC migration was investigated by wound‑healing assay following overexpression and knockdown. Functional enrichment analysis of the whole‑genome expression profiles of PASMCs with EZH2 overexpression was performed using an mRNA Human Gene Expression Microarray. Quantitative (q)PCR was performed to confirm the results of the microarray. EZH2 expression levels increased in CTEPH cell models. The overexpression of EZH2 enhanced PASMC migration compared with control conditions. Functional enrichment analysis of the differentially expressed genes following EZH2 overexpression indicated a strong link between EZH2 and the immune inflammatory response and oxidoreductase activity in PASMCs. mRNA expression levels of superoxide dismutase 3 were verified by qPCR. The results suggested that EZH2 was involved in the migration of PASMCs in PH, and may serve as a potential target for the treatment of PH.
Collapse
Affiliation(s)
- Ying Wang
- Department of Clinical Laboratory, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Xiao-Xi Huang
- Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Institute of Respiratory Medicine, Beijing 100020, P.R. China
| | - Dong Leng
- Department of Clinical Laboratory, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Ji-Feng Li
- Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Institute of Respiratory Medicine, Beijing 100020, P.R. China
| | - Yan Liang
- Department of Clinical Laboratory, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| |
Collapse
|
11
|
Arthur Ataam J, Amsallem M, Guihaire J, Haddad F, Lamrani L, Stephan F, Jaïs X, Humbert M, Mercier O, Fadel E. Preoperative C-reactive protein predicts early postoperative outcomes after pulmonary endarterectomy in patients with chronic thromboembolic pulmonary hypertension. J Thorac Cardiovasc Surg 2020; 161:1532-1542.e5. [PMID: 32007254 DOI: 10.1016/j.jtcvs.2019.11.133] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 11/09/2019] [Accepted: 11/27/2019] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To determine whether preoperative systemic inflammation (defined by C-reactive protein [CRP] levels ≥10 mg/L) is associated with worse functional and hemodynamic status and poor early outcomes postendarterectomy in patients with chronic thromboembolic pulmonary hypertension (CTEPH). METHODS This study included 159 patients who underwent pulmonary endarterectomy from 2009 to 2013 (derivation cohort) and 238 patients from 2015 to 2016 (validation cohort) with CRP data from the national CTEPH registry. The correlations between proinflammatory markers (CRP, interleukins 1 and 6, fibrinogen, and leukocytes) and hemodynamics were assessed in the derivation cohort. Pre-, perioperative characteristics, and 30-day outcomes (ie, death or lung transplant or extracorporeal membrane oxygenation need or inotropic or vasopressor need ≥3 days) of patients with CRP levels ≥ or <10 mg/L were compared. RESULTS Median age of the derivation cohort was 63 [52-73] years with 48% female, 80% in New York Heart Association class III/IV. The validation cohort had similar demographics and disease severity. Patients with CRP ≥10 mg/L had greater resistance levels and lower cardiac index than those with CRP <10 mg/L in both cohorts. The primary endpoint was reached in 38% (derivation) and 42% (validation) of patients. In multivariable logistic regression analysis, CRP ≥10 mg/L was associated with the primary endpoint in both the derivation cohort (odd ratio, 2.49 [1.11-5.61], independently of New York Heart class class IV and aortic clamping duration) and the validation cohort (odd ratio, 1.89 [1.09-3.61], independently of age and aortic clamping duration). CONCLUSIONS Preoperative CRP ≥10 mg/L is independently associated with adverse early outcomes postendarterectomy.
Collapse
Affiliation(s)
- Jennifer Arthur Ataam
- Research and Innovation Unit, INSERM U999, DHU Torino, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France; Department of Cardiovascular Medicine, Stanford Cardiovascular Institute, Stanford, Calif
| | - Myriam Amsallem
- Research and Innovation Unit, INSERM U999, DHU Torino, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France; Department of Cardiovascular Medicine, Stanford Cardiovascular Institute, Stanford, Calif.
| | - Julien Guihaire
- Research and Innovation Unit, INSERM U999, DHU Torino, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France; Department of Cardiothoracic Surgery, Marie Lannelongue Hospital, Le Plessis Robinson, France
| | - Francois Haddad
- Department of Cardiovascular Medicine, Stanford Cardiovascular Institute, Stanford, Calif
| | - Lilia Lamrani
- Research and Innovation Unit, INSERM U999, DHU Torino, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France
| | - Francois Stephan
- Department of Intensive Care Unit, Marie Lannelongue Hospital, Le Plessis Robinson, France
| | - Xavier Jaïs
- Department of Pulmonary Diseases, Kremlin Bicetre Hospital‒APHP, Kremlin Bicetre, France
| | - Marc Humbert
- Department of Pulmonary Diseases, Kremlin Bicetre Hospital‒APHP, Kremlin Bicetre, France
| | - Olaf Mercier
- Research and Innovation Unit, INSERM U999, DHU Torino, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France; Department of Cardiothoracic Surgery, Marie Lannelongue Hospital, Le Plessis Robinson, France
| | - Elie Fadel
- Research and Innovation Unit, INSERM U999, DHU Torino, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France; Department of Cardiothoracic Surgery, Marie Lannelongue Hospital, Le Plessis Robinson, France
| |
Collapse
|
12
|
Naito A, Sakao S, Lang IM, Voelkel NF, Jujo T, Ishida K, Sugiura T, Matsumiya G, Yoshino I, Tanabe N, Tatsumi K. Endothelial cells from pulmonary endarterectomy specimens possess a high angiogenic potential and express high levels of hepatocyte growth factor. BMC Pulm Med 2018; 18:197. [PMID: 30594174 PMCID: PMC6310963 DOI: 10.1186/s12890-018-0769-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 12/18/2018] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Impaired angiogenesis is assumed to be an important factor in the development of chronic thromboembolic pulmonary hypertension (CTEPH). However, the role of endothelial cells (ECs) in CTEPH remains unclear. The aim of this study was to investigate the angiogenic potential of ECs from pulmonary endarterectomy (PEA) specimens. METHODS We isolated ECs from PEA specimens (CTEPH-ECs) and control EC lines from the intact pulmonary arteries of patients with peripheral lung cancers, using a MACS system. These cells were analyzed in vitro including PCR-array analysis, and the PEA specimens were analyzed with immunohistochemistry. Additionally, the serum HGF levels were determined in CTEPH patients. RESULTS A three-dimensional culture assay revealed that CTEPH-ECs were highly angiogenic. An angiogenesis-focused gene PCR array revealed a high expression of hepatocyte growth factor (HGF) in CTEPH-ECs. The high expression of HGF was also confirmed in the supernatant extracted from PEA specimens. The immunohistochemical analysis showed expression of HGF on the surface of the thrombus vessels. The serum HGF levels in CTEPH patients were higher than those in pulmonary thromboembolism survivors. CONCLUSION Our study suggests that there are ECs with pro-angiogenetic character and high expression of HGF in PEA specimens. It remains unknown how these results are attributable to the etiology. However, further investigation focused on the HGF pathway may provide novel diagnostic and therapeutic tools for patients with CTEPH.
Collapse
Affiliation(s)
- Akira Naito
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan.,Department of Advancing Research on Treatment Strategies for respiratory disease, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan.
| | - Irene M Lang
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Norbert F Voelkel
- Victoria Johnson Center for Obstructive Lung Disease, Virginia Commonwealth University, 1101 East Marshall Street, Sanger Hall, Richmond, VA, 23298-0565, USA
| | - Takayuki Jujo
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan.,Department of Advanced Medicine in Pulmonary Hypertension, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Keiichi Ishida
- Department of Cardiovascular Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Toshihiko Sugiura
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Goro Matsumiya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Ichiro Yoshino
- Department of Thoracic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Nobuhiro Tanabe
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba City, 260-8670, Japan
| |
Collapse
|
13
|
Sydykov A, Mamazhakypov A, Petrovic A, Kosanovic D, Sarybaev AS, Weissmann N, Ghofrani HA, Schermuly RT. Inflammatory Mediators Drive Adverse Right Ventricular Remodeling and Dysfunction and Serve as Potential Biomarkers. Front Physiol 2018; 9:609. [PMID: 29875701 PMCID: PMC5974151 DOI: 10.3389/fphys.2018.00609] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 05/04/2018] [Indexed: 01/07/2023] Open
Abstract
Adverse right ventricular (RV) remodeling leads to ventricular dysfunction and failure that represents an important determinant of outcome in patients with pulmonary hypertension (PH). Recent evidence indicates that inflammatory activation contributes to the pathogenesis of adverse RV remodeling and dysfunction. It has been shown that accumulation of inflammatory cells such as macrophages and mast cells in the right ventricle is associated with maladaptive RV remodeling. In addition, inhibition of inflammation in animal models of RV failure ameliorated RV structural and functional impairment. Furthermore, a number of circulating inflammatory mediators have been demonstrated to be associated with RV performance. This work reviews the role of inflammation in RV remodeling and dysfunction and discusses anti-inflammatory strategies that may attenuate adverse structural alterations while promoting improvement of RV function.
Collapse
Affiliation(s)
- Akylbek Sydykov
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany.,Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek, Kyrgyzstan
| | - Argen Mamazhakypov
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Aleksandar Petrovic
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Djuro Kosanovic
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Akpay S Sarybaev
- Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek, Kyrgyzstan
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Hossein A Ghofrani
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Ralph T Schermuly
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| |
Collapse
|
14
|
Wang J, Zhao H, Wang Y, Herrmann HC, Witschey WRT, Han Y. Native T1 and T2 mapping by cardiovascular magnetic resonance imaging in pressure overloaded left and right heart diseases. J Thorac Dis 2018; 10:2968-2975. [PMID: 29997963 DOI: 10.21037/jtd.2018.04.141] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Background Pulmonary arterial hypertension (PAH) and severe aortic valve stenosis (AS) are diseases characterized by increased afterload of the right and left heart, respectively. Our study aims to investigate the differences of myocardial tissue characteristics in the pressure overloaded left and right hearts, especially in the shared interventricular septum, as detected by native T1 and T2 relaxation times. Methods Eighteen patients with PAH and 19 patients with severe AS in addition to 5 healthy volunteers underwent 1.5-T CMR examination with native T1 and T2 mapping. Mean T1 and T2 value were measured at the right ventricular (RV) free wall, superior RV insertion, inferior RV insertion, interventricular septum and left ventricular (LV) lateral wall. Results Compared with controls and AS group, T1 was significantly elevated in the RV insertion in PAH group (P=0.015), while no statistically significant differences were seen in other segments among the three groups. There was an increase of T2 in the RV insertion in AS and PAH groups (P=0.01). Significant T2 elevation was also observed in the RV free wall of PAH group, and the LV lateral wall of AS group compared with the control group. RV insertion T2 was significantly correlated with RV end-diastolic volume index (r=0.608, P=0.016) and RV mass index (r=0.57, P=0.026) in the PAH group. LV lateral wall T2 and RV insertion T2 were significantly correlated with aortic valve mean gradients in the AS group (r=0.56, P=0.02; r=0.58, P=0.01, respectively). Conclusions In pressure overload diseases, both T1 and T2 values increase in the myocardium. The alterations seen in the RV insertion sites in the septum was more pronounced with RV pressure overload. T2 values also correlated with structural and functional remodeling in both diseases. Combining T1 and T2 mapping may help to better characterize the alternation of myocardial composition in pressure overloaded heart diseases.
Collapse
Affiliation(s)
- Jing Wang
- Cardiovascular Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Cardiology, PLA General Hospital, Beijing 100853, China
| | - Hang Zhao
- Cardiovascular Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Cardiology, Shanghai Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
| | - Yue Wang
- Cardiovascular Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Howard C Herrmann
- Cardiovascular Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Walter R T Witschey
- Department of Radiology, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA
| | - Yuchi Han
- Cardiovascular Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
15
|
Mohammadifard N, Humphries KH, Gotay C, Mena-Sánchez G, Salas-Salvadó J, Esmaillzadeh A, Ignaszewski A, Sarrafzadegan N. Trace minerals intake: Risks and benefits for cardiovascular health. Crit Rev Food Sci Nutr 2017; 59:1334-1346. [PMID: 29236516 DOI: 10.1080/10408398.2017.1406332] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Minerals play a major role in regulating cardiovascular function. Imbalances in electrolyte minerals are frequent and potentially hazardous occurrences that may lead to the development of cardiovascular diseases (CVDs). Transition metals, such as iron, zinc, copper and selenium, play a major role in cell metabolism. However, there is controversy over the effects of dietary and supplemental intake of these metals on cardiovascular risk factors and events. Since their pro-oxidant or antioxidant functions can have different effects on cardiovascular health. While deficiency of these trace elements can cause cardiovascular dysfunction, several studies have also shown a positive association between metal serum levels and cardiovascular risk factors and events. Thus, a J- or U-shaped relationship between the transition minerals and cardiovascular events has been proposed. Given the existing controversies, large, well-designed, long-term, randomized clinical trials are required to better examine the effects of trace mineral intake on cardiovascular events and all-cause mortality in the general population. In this review, we discuss the role of dietary and/or supplemental iron, copper, zinc, and selenium on cardiovascular health. We will also clarify their clinical applications, benefits, and harms in CVDs prevention.
Collapse
Affiliation(s)
- Noushin Mohammadifard
- a Hypertension Research Center , Cardiovascular Research Institute, Isfahan University of Medical Sciences , Isfahan , Iran
- b Interventional Cardiology Research Center , Cardiovascular Research Institute, Isfahan University of Medical Sciences , Isfahan , Iran
| | - Karin H Humphries
- c Women's Cardiovascular Health , Department of Medicine, The University of British Columbia , Vancouver , Canada
| | - Carolyn Gotay
- d Centre of Excellence in Cancer Prevention, Faculty of Medicine, School of Population and Public Health, The University of British Columbia , Vancouver , Canada
| | - Guillermo Mena-Sánchez
- e Human Nutrition Unit , Department of Biochemistry & Biotechnology , IISPV, School of Medicine, Rovira i Virgili University, and CIBER Obesity and Nutrition , Reus , Spain
| | - Jordi Salas-Salvadó
- e Human Nutrition Unit , Department of Biochemistry & Biotechnology , IISPV, School of Medicine, Rovira i Virgili University, and CIBER Obesity and Nutrition , Reus , Spain
| | - Ahmad Esmaillzadeh
- f Obesity and Eating Habits Research Center , Endocrinology and Metabolism Molecular, Cellular Sciences Institute, Tehran University of Medical Sciences , Tehran , Iran
- g Department of Community Nutrition , School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences , Tehran , Iran
- h Department of Community Nutrition , School of Nutrition and Food Science, Isfahan University of Medical Sciences , Isfahan , Iran
| | - Andrew Ignaszewski
- i Division of Cardiology, Faculty of Medicine, The University of British Columbia , Vancouver , Canada
| | - Nizal Sarrafzadegan
- j Isfahan Cardiovascular Research Center , Cardiovascular Research Institute, Isfahan University of Medical Sciences , Isfahan , Iran
| |
Collapse
|
16
|
Mercier O, Arthur Ataam J, Langer NB, Dorfmüller P, Lamrani L, Lecerf F, Decante B, Dartevelle P, Eddahibi S, Fadel E. Abnormal pulmonary endothelial cells may underlie the enigmatic pathogenesis of chronic thromboembolic pulmonary hypertension. J Heart Lung Transplant 2016; 36:305-314. [PMID: 27793518 DOI: 10.1016/j.healun.2016.08.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 06/30/2016] [Accepted: 08/17/2016] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Chronic thromboembolic pulmonary hypertension results from chronic mechanical obstruction of the pulmonary arteries after acute venous thromboembolism. However, the mechanisms that result in the progression from unresolved thrombus to fibrotic vascular remodeling are unknown. We hypothesized that pulmonary artery endothelial cells contribute to this phenomenon via paracrine growth factor and cytokine signaling. METHODS Using enzyme-linked immunosorbent assay and cell migration assays, we investigated the circulating growth factors and cytokines of chronic thromboembolic pulmonary hypertension patients as well as the cross talk between pulmonary endothelial cells and pulmonary artery smooth muscle cells and monocytes from patients with chronic thromboembolic pulmonary hypertension in vitro. RESULTS Culture medium from the pulmonary endothelial cells of chronic thromboembolic pulmonary hypertension patients contained higher levels of growth factors (fibroblast growth factor 2), inflammatory cytokines (interleukin 1β, interleukin 6, monocyte chemoattractant protein 1), and cell adhesion molecules (vascular cell adhesion molecule 1 and intercellular adhesion molecule 1). Furthermore, exposure to the culture medium of pulmonary endothelial cells from patients with chronic thromboembolic pulmonary hypertension elicited marked pulmonary artery smooth muscle cell growth and monocyte migration. CONCLUSIONS These findings implicate pulmonary endothelial cells as key regulators of pulmonary artery smooth muscle cell and monocyte behavior in chronic thromboembolic pulmonary hypertension and suggest a potential mechanism for the progression from unresolved thrombus to fibrotic vascular remodeling.
Collapse
Affiliation(s)
- Olaf Mercier
- Research and Innovation Unit, INSERM U999, DHU TORINO, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France; Departments of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, Le Plessis Robinson, France.
| | - Jennifer Arthur Ataam
- Research and Innovation Unit, INSERM U999, DHU TORINO, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France
| | - Nathaniel B Langer
- Research and Innovation Unit, INSERM U999, DHU TORINO, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France; Departments of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, Le Plessis Robinson, France
| | - Peter Dorfmüller
- Research and Innovation Unit, INSERM U999, DHU TORINO, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France; Pathology, Marie Lannelongue Hospital, Le Plessis Robinson, France
| | - Lilia Lamrani
- Research and Innovation Unit, INSERM U999, DHU TORINO, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France
| | - Florence Lecerf
- Research and Innovation Unit, INSERM U999, DHU TORINO, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France
| | - Benoit Decante
- Research and Innovation Unit, INSERM U999, DHU TORINO, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France
| | - Philippe Dartevelle
- Research and Innovation Unit, INSERM U999, DHU TORINO, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France; Departments of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, Le Plessis Robinson, France
| | - Saadia Eddahibi
- Research and Innovation Unit, INSERM U999, DHU TORINO, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France; INSERM U1046, CNRS UMR 9214, Université de Montpellier, CHU Arnaud de Villeneuve Montpellier, Montpellier, France
| | - Elie Fadel
- Research and Innovation Unit, INSERM U999, DHU TORINO, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France; Departments of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, Le Plessis Robinson, France
| |
Collapse
|
17
|
The Clinical Significance of HbA1c in Operable Chronic Thromboembolic Pulmonary Hypertension. PLoS One 2016; 11:e0152580. [PMID: 27031508 PMCID: PMC4816563 DOI: 10.1371/journal.pone.0152580] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 03/16/2016] [Indexed: 01/30/2023] Open
Abstract
Background Glycosylated hemoglobin A1c (HbA1c) has been proposed as an independent predictor of long-term prognosis in pulmonary arterial hypertension. However, the clinical relevance of HbA1c in patients with operable chronic thromboembolic pulmonary hypertension (CTEPH) remains unknown. The aim of the present study was to investigate the clinical significance of HbA1c as a biomarker in CTEPH. Methods Prospectively, 102 patients underwent pulmonary endarterectomy (PEA) in our national referral center between March 2013 and March 2014, of which after exclusion 45 patients were analyzed. HbA1c- levels, hemodynamic and exercise parameters were analyzed prior and one-year post-PEA. Results 45 patients (BMI: 27.3 ± 6.0 kg/m2; age: 62.7 ± 12.3 years) with a mean pulmonary arterial pressure (mPAP) of 43.6 ± 9.4 mmHg, a pulmonary vascular resistance (PVR) of 712.1 ± 520.4 dyn*s/cm5, a cardiac index (CI) of 2.4 ± 0.5 l/min/m2 and a mean HbA1c-level of 39.8 ± 5.6 mmol/mol were included. One-year post-PEA pulmonary hemodynamic and functional status significantly improved in our cohort. Baseline HbA1c-levels were significantly associated with CI, right atrial pressure, peak oxygen uptake and the change of 6-minute walking distance using linear regression analysis. However, using logistic regression analysis baseline HbA1c-levels were not significantly associated with residual post-PEA PH. Conclusions This is the first prospective study to describe an association of HbA1c-levels with pulmonary hemodynamics and exercise capacity in operable CTEPH patients. Our preliminary results indicate that in these patients impaired glucose metabolism as assessed by HbA1c is of clinical significance. However, HbA1c failed as a predictor of the hemodynamic outcome one-year post-PEA.
Collapse
|
18
|
Fonseca S, Mendonça V, Teles M, Ribeiro V, Tossige-Gomes R, Neves C, Rocha-Vieira E, Leite L, Soares D, Coimbra C, Lacerda A. Inflammatory cytokines and plasma redox status responses in hypertensive subjects after heat exposure. Braz J Med Biol Res 2016; 49:S0100-879X2016000300701. [PMID: 26840715 PMCID: PMC4763820 DOI: 10.1590/1414-431x20155026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/28/2015] [Indexed: 02/07/2023] Open
Abstract
Hypertension is characterized by a pro-inflammatory status, including redox imbalance and increased levels of pro-inflammatory cytokines, which may be exacerbated after heat exposure. However, the effects of heat exposure, specifically in individuals with inflammatory chronic diseases such as hypertension, are complex and not well understood. This study compared the effects of heat exposure on plasma cytokine levels and redox status parameters in 8 hypertensive (H) and 8 normotensive (N) subjects (age: 46.5±1.3 and 45.6±1.4 years old, body mass index: 25.8±0.8 and 25.6±0.6 kg/m2, mean arterial pressure: 98.0±2.8 and 86.0±2.3 mmHg, respectively). They remained at rest in a sitting position for 10 min in a thermoneutral environment (22°C) followed by 30 min in a heated environmental chamber (38°C and 60% relative humidity). Blood samples were collected before and after heat exposure. Plasma cytokine levels were measured using sandwich ELISA kits. Plasma redox status was determined by thiobarbituric acid reactive substances (TBARS) levels and ferric reducing ability of plasma (FRAP). Hypertensive subjects showed higher plasma levels of IL-10 at baseline (P<0.05), although levels of this cytokine were similar between groups after heat exposure. Moreover, after heat exposure, hypertensive individuals showed higher plasma levels of soluble TNF receptor (sTNFR1) and lower TBARS (P<0.01) and FRAP (P<0.05) levels. Controlled hypertensive subjects, who use angiotensin-converting-enzyme inhibitor (ACE inhibitors), present an anti-inflammatory status and balanced redox status. Nevertheless, exposure to a heat stress condition seems to cause an imbalance in the redox status and an unregulated inflammatory response.
Collapse
Affiliation(s)
- S.F. Fonseca
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde, Universidade
Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
- Programa Multicêntrico de Pós Graduação em Ciências Fisiológicas,
Sociedade Brasileira de Fisiologia, São Paulo, SP, Brasil
| | - V.A. Mendonça
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde, Universidade
Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
- Programa Multicêntrico de Pós Graduação em Ciências Fisiológicas,
Sociedade Brasileira de Fisiologia, São Paulo, SP, Brasil
| | - M.C. Teles
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde, Universidade
Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
- Programa Multicêntrico de Pós Graduação em Ciências Fisiológicas,
Sociedade Brasileira de Fisiologia, São Paulo, SP, Brasil
| | - V.G.C. Ribeiro
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde, Universidade
Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
- Programa Multicêntrico de Pós Graduação em Ciências Fisiológicas,
Sociedade Brasileira de Fisiologia, São Paulo, SP, Brasil
| | - R. Tossige-Gomes
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde, Universidade
Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
- Programa Multicêntrico de Pós Graduação em Ciências Fisiológicas,
Sociedade Brasileira de Fisiologia, São Paulo, SP, Brasil
| | - C.D.C. Neves
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde, Universidade
Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
- Programa Multicêntrico de Pós Graduação em Ciências Fisiológicas,
Sociedade Brasileira de Fisiologia, São Paulo, SP, Brasil
| | - E. Rocha-Vieira
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde, Universidade
Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
- Programa Multicêntrico de Pós Graduação em Ciências Fisiológicas,
Sociedade Brasileira de Fisiologia, São Paulo, SP, Brasil
| | - L.H.R. Leite
- Instituto de Ciências Biológicas, Universidade Federal de Juiz de
Fora, Juiz de Fora, MG, Brasil
| | - D.D. Soares
- Escola de Educação Física, Universidade Federal de Minas Gerais, Belo
Horizonte, MG, Brasil
| | - C.C. Coimbra
- Instituto de Ciências Biológicas, Universidade Federal de Minas
Gerais, Belo Horizonte, MG, Brasil
| | - A.C.R. Lacerda
- Centro Integrado de Pós-Graduação e Pesquisa em Saúde, Universidade
Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
- Programa Multicêntrico de Pós Graduação em Ciências Fisiológicas,
Sociedade Brasileira de Fisiologia, São Paulo, SP, Brasil
| |
Collapse
|
19
|
Saghazadeh A, Rezaei N. Inflammation as a cause of venous thromboembolism. Crit Rev Oncol Hematol 2016; 99:272-85. [DOI: 10.1016/j.critrevonc.2016.01.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 11/27/2015] [Accepted: 01/12/2016] [Indexed: 12/12/2022] Open
|
20
|
Wang Y, Hu F, Mu X, Wu F, Yang D, Zheng G, Sun X, Gong K, Zhang Z. Protective effects of drag-reducing polymers in a rat model of monocrotaline-induced pulmonary hypertension. Biorheology 2016; 53:13-22. [PMID: 26889655 DOI: 10.3233/bir-15062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Drag-reducing polymers (DRPs) are blood-soluble macromolecules which may increase blood flow and reduce vascular resistance. The purpose of the present study was to observe the effect of DRPs on monocrotaline-induced pulmonary hypertension (PH) in the rat model. METHODS A total of 64 male Wistar rats were randomly divided into four groups: Group I (pulmonary hypertension model + DRP treatment); Group II (pulmonary hypertension model + saline treatment); Group III (control + DRP treatment); Group IV (control + saline treatment). After five weeks, comparisons were made of the following indices: survival rate, body weight, blood pressure, right ventricular systolic pressure, right ventricular hypertrophy, wall thickness of pulmonary arteries, the internal diameter of small pulmonary arteries, plasma IL-1β and IL-6. RESULTS The survival rate after 5 weeks varied significantly across all groups (P=0.013), but the survival rates of Groups I and II were not statistically significantly different. Administration of DRP (intravenous injection twice weekly) attenuated the PH-induced increase in right ventricular systolic pressure and suppressed the increases in right ventricular (RV) weight and the ratio of right ventricular weight to left ventricle plus septum weight (RV/LV + S). DRP treatment also significantly decreased the wall thickness of pulmonary arteries, augmented the internal diameter of small pulmonary arteries, and suppressed increases in the plasma levels of IL-1β and IL-6. CONCLUSIONS DRP treatment with intravenous injection effectively inhibited the development of monocrotaline-induced pulmonary hypertension in the rat model. DRPs may have potential application for the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
- Yali Wang
- Department of Respiratory Diseases, The Second Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Feng Hu
- Department of Cardiovascular Diseases, The Second Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Xiaoyan Mu
- Department of Respiratory Diseases, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Feng Wu
- Department of Respiratory Diseases, The Second Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Dechun Yang
- Department of Respiratory Diseases, The Second Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Guixiang Zheng
- Department of Cardiovascular Diseases, The Second Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Xiaoning Sun
- Department of Cardiovascular Diseases, The Second Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Kaizheng Gong
- Department of Cardiovascular Diseases, The Second Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Zhengang Zhang
- Department of Cardiovascular Diseases, The Second Clinical Medical College, Yangzhou University, Yangzhou, China
| |
Collapse
|
21
|
von Haehling S, Jankowska EA, van Veldhuisen DJ, Ponikowski P, Anker SD. Iron deficiency and cardiovascular disease. Nat Rev Cardiol 2015; 12:659-69. [DOI: 10.1038/nrcardio.2015.109] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
22
|
Poels EM, da Costa Martins PA, van Empel VPM. Adaptive capacity of the right ventricle: why does it fail? Am J Physiol Heart Circ Physiol 2015; 308:H803-13. [DOI: 10.1152/ajpheart.00573.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 01/21/2015] [Indexed: 11/22/2022]
Abstract
Only in recent years has the right ventricle (RV) function become appreciated to be equally important to the left ventricle (LV) function to maintain cardiac output. Right ventricular failure is, irrespectively of the etiology, associated with impaired exercise tolerance and poor survival. Since the anatomy and physiology of the RV is distinctly different than that of the LV, its adaptive mechanisms and the pathways involved are different as well. RV hypertrophy is an important mechanism of the RV to preserve cardiac output. This review summarizes the current knowledge on the right ventricle and its response to pathologic situations. We will focus on the adaptive capacity of the right ventricle and the molecular pathways involved, and we will discuss potential therapeutic interventions.
Collapse
Affiliation(s)
- Ella M. Poels
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands; and
- Department of Cardiology, Heart Vessel Center, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Paula A. da Costa Martins
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands; and
| | - Vanessa P. M. van Empel
- Department of Cardiology, Heart Vessel Center, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
23
|
Nazemiyeh M, Sharifi A, Amiran F, Pourafkari L, Taban Sadeghi M, Namdar H, Abbasnezhad M. Relationship Between Prohormone Brain Natriuretic Peptide (NT-proBNP) Level and Severity of Pulmonary Dysfunction in Patients With Chronic Congestive Heart Failure. J Cardiovasc Thorac Res 2015; 7:24-7. [PMID: 25859312 PMCID: PMC4378671 DOI: 10.15171/jcvtr.2015.05] [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: 01/10/2015] [Accepted: 02/14/2015] [Indexed: 11/22/2022] Open
Abstract
Introduction: Congestive heart failure (CHF) is a common disease and its prevalence is increasing in industrialized countries. NT-proBNP measurement is an established diagnostic test for diagnosis of CHF in patients who present to emergency room with acute dyspnea. The primary object of this study was to determine the relationship between levels of brain natriuretic peptide precursor and severity of lung function impairment in patients with chronic CHF.
Methods: This cross-sectional and analytical study that performed in Tuberculosis and Lung Disease Research Center of Tabriz University of Medical Sciences on 95 patients with chronic heart failure, and relation between NT-proBNP levels and pulmonary function parameters were examined.
Results: Sixty-four patients were male and 31 were female. The average age of male and females was 62.90 ± 11.54 and 61.61 ± 11.98 years, respectively. A significant inverse linear correlation was found between NT-proBNP and FEV1 (P < 0.001, r = -0.367), FVC (P < 0.001, r = -0.444), TLC (P = 0.022, r = -0.238), maximal midexpiratory flow (MMEF) (P = 0.047, r = -0.207) and left ventricular ejection fraction (LVEF) (P < 0.001, r = -0.461). A significant positive linear correlation was found between NT-proBNP and FEV1/FVC (P = 0.013, r = 0.257), RV/TLC (P = 0.003, r=0.303) and 5 Hz Raw (r = 0.231, P = 0.024).
Conclusion: This study showed that, both restrictive and obstructive ventilator impairments can occur in chronic CHF and as NT-proBNP increases appropriate to hemodynamic deterioration, pulmonary dysfunction increases.
Collapse
Affiliation(s)
- Masoud Nazemiyeh
- Tuberculosis and Lung Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Akbar Sharifi
- Tuberculosis and Lung Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Amiran
- Department of Internal Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili Pourafkari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hossein Namdar
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Abbasnezhad
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
24
|
Chronic thromboembolic pulmonary hypertension is not associated with iron overload. Cardiovasc Pathol 2014; 24:76-9. [PMID: 25449047 DOI: 10.1016/j.carpath.2014.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 09/08/2014] [Accepted: 09/22/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Chronic thromboembolic pulmonary hypertension (CTEPH) is characterized as the incomplete resolution of emboli after pulmonary embolism (PE) and the subsequent fibrotic organization and remodeling of pulmonary vascular bed. It has been reported that abnormal fibrin probably contributes to the incomplete resolution of emboli. And there is evidence that free iron could convert fibrinogen into fibrin which is remarkably resistant to lysis. Thus, we hypothesized that persistent iron overload might participate in the development of CTEPH. MATERIALS AND METHODS A case-control study was conducted. Forty-five CTEPH patients were enrolled as cases, and 36 age and sex frequency-matched chronic PE patients without pulmonary hypertension were selected as controls. Levels of free iron, soluble transferrin receptor (sTfR), ferritin, sTfR/ferritin ratio, hepcidin-25, tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, and malondialdehyde (MDA) were compared between the two groups. Logistic regression analysis was carried out to estimate odds ratios. RESULTS There was no difference of the levels of free iron, hepcidin-25, sTfR, ferritin, sTfR/ferritin ratio, TNF-α, and MDA between CTEPH patients and the controls. Levels of sTfR and ferritin in both groups were within the normal limits. Levels of IL-6 in CTEPH patients were significantly higher than that in the controls. A negative correlation was observed between hepcidin-25 and sTfR (Spearman's r=-0.438, P<.001), and a positive correlation was observed between hepcidin-25 and ferritin (Spearman's r=0.503, P<.001). In the univariate logistic regression model, there was no association observed between CTEPH and free iron, hepcidin-25, sTfR, ferritin, sTfR/ferritin ratio, TNF-α, IL-6, and MDA. CONCLUSIONS CTEPH has no association with iron overload. The iron status evaluated by sTfR and ferritin is within the normal limits in this CTEPH population.
Collapse
|
25
|
Local and systemic RAGE axis changes in pulmonary hypertension: CTEPH and iPAH. PLoS One 2014; 9:e106440. [PMID: 25188497 PMCID: PMC4154707 DOI: 10.1371/journal.pone.0106440] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 07/29/2014] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE The molecular determinants of chronic thromboembolic pulmonary hypertension (CTEPH) and idiopathic pulmonary arterial hypertension (iPAH) remain poorly understood. The receptor for advanced glycation endproducts (RAGE) and its ligands: HMGB1 and S100A9 are involved in inflammatory disorders. We sought to investigate the role of the RAGE axis in patients with CTEPH undergoing pulmonary endarterectomy (PEA), iPAH undergoing lung transplantation (LuTX). The high pulmonary vascular resistance in CTEPH/iPAH results in pressure overload of the right ventricle. We compared sRAGE measurements to that of patients with aortic valve stenosis (AVS) - pressure overload of the left ventricle. METHODS We enrolled patients with CTEPH(26), iPAH(15), AVS(15) and volunteers(33). Immunohistochemistry with antibodies to RAGE and HMGB1 was performed on PEA specimens and lung tissues. We employed enzyme-linked immunosorbent assays to determine the concentrations of sRAGE, esRAGE, HMGB1 and S100A9 in serum of volunteers and patients with CTEPH, iPAH, AVS before and after PEA, LuTX and aortic valve replacement (AVR). RESULTS In endarterectomised tissues from patients with CTEPH RAGE and HMGB1 were identified in myofibroblasts (α-SMA+vimentin+CD34-), recanalizing vessel-like structures of distal myofibrotic tissues and endothelium of neointima. RAGE was differentially expressed in prototypical Heath Edwards lesions in iPAH. We found significantly increased serum concentrations of sRAGE, esRAGE and HMGB1 in CTEPH. In iPAH, sRAGE and esRAGE were significantly higher than in controls. Serum concentrations of sRAGE were significantly elevated in iPAH(p<0.001) and CTEPH(p = 0.001) compared to AVS. Serum sRAGE was significantly higher in iPAH compared to CTEPH(p = 0.042) and significantly reduced in AVS compared to controls(p = 0.001). There were no significant differences in sRAGE serum concentrations before and after surgical therapy for CTEPH, iPAH or AVS. CONCLUSIONS Our data suggest a role for the RAGE pathway in the pathophysiology of CTEPH and iPAH. PEA improves the local control of disease but may not influence the systemic inflammatory mechanisms in CTEPH patients through the RAGE pathway.
Collapse
|
26
|
Zabini D, Heinemann A, Foris V, Nagaraj C, Nierlich P, Bálint Z, Kwapiszewska G, Lang IM, Klepetko W, Olschewski H, Olschewski A. Comprehensive analysis of inflammatory markers in chronic thromboembolic pulmonary hypertension patients. Eur Respir J 2014; 44:951-62. [PMID: 25034560 DOI: 10.1183/09031936.00145013] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is associated with chronic inflammation but the pathological mechanisms are largely unknown. Our study aimed to simultaneously profile a broad range of cytokines in the supernatant of pulmonary endarterectomy (PEA) surgical material, as well as prospectively in patients with CTEPH to investigate whether circulating cytokines are associated with haemodynamic and physical characteristics of CTEPH patients. Herein, we show that PEA specimens revealed a significant upregulation of interleukin (IL)-6, monocyte chemoattractant protein-1, interferon-γ-induced protein-10 (IP)-10, macrophage inflammatory protein (MIP)1α and RANTES compared to lung tissue from healthy controls. In prospectively collected serum, levels of IL-6, IL-8, IP-10, monokine induced by interferon-γ (MIG) and MIP1α were significantly elevated in CTEPH patients compared to age- and sex-matched healthy controls. In serum of idiopathic pulmonary arterial hypertension (IPAH) patients, only IP-10 and MIG were significantly increased. In CTEPH but not in IPAH, IP-10 was negatively correlated with cardiac index, 6-min walking distance and carbon monoxide diffusion capacity. In vitro, IP-10 significantly increased migration of freshly isolated adventitial fibroblasts. Our study is the first to show that IP-10 secretion is associated with poor pulmonary haemodynamics and physical capacity in CTEPH and might be involved in the pathological mechanism of PEA tissue formation.
Collapse
Affiliation(s)
- Diana Zabini
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Akos Heinemann
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Vasile Foris
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Patrick Nierlich
- Dept of Surgery, Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Zoltán Bálint
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria Dept of Anaesthesia and Intensive Care Medicine, Experimental Anaesthesiology, Medical University of Graz, Graz, Austria
| | - Irene M Lang
- Dept of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria
| | - Walter Klepetko
- Dept of Surgery, Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Horst Olschewski
- Dept of Internal Medicine, Division of Pulmonology, Medical University of Graz, Graz, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria Dept of Anaesthesia and Intensive Care Medicine, Experimental Anaesthesiology, Medical University of Graz, Graz, Austria
| |
Collapse
|
27
|
Yang T, Li ZN, Chen G, Gu Q, Ni XH, Zhao ZH, Ye J, Meng XM, Liu ZH, Xiong CM, He JG. Increased levels of plasma CXC-Chemokine Ligand 10, 12 and 16 are associated with right ventricular function in patients with idiopathic pulmonary arterial hypertension. Heart Lung 2014; 43:322-7. [PMID: 24856224 DOI: 10.1016/j.hrtlng.2014.04.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 04/16/2014] [Accepted: 04/17/2014] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To investigate plasma levels of CXC-Chemokine Ligand 10 (CXCL10), CXC-Chemokine Ligand 12 (CXCL12) and CXC-Chemokine Ligand 16 (CXCL16) in patients with idiopathic pulmonary arterial hypertension (IPAH). METHODS Plasma levels of biomarkers were measured by enzyme-linked immunosorbent assay in 61 patients with IPAH and 20 healthy volunteers. RESULTS Plasma CXCL10, CXCL12 and CXCL16 concentrations were increased significantly in IPAH patients compared with controls, and significantly correlated with N-terminal pro-brain natriuretic peptide, tricuspid annulus plane systolic excursion and right ventricular ejection fraction. CONCLUSIONS Increased levels of CXCL10, CXCL12 and CXCL16 are associated with right ventricular dysfunction in patients with IPAH.
Collapse
Affiliation(s)
- Tao Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beilishi Road, Xicheng District, Beijing 100037, China
| | - Zhen-Nan Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beilishi Road, Xicheng District, Beijing 100037, China
| | - Guo Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beilishi Road, Xicheng District, Beijing 100037, China
| | - Qing Gu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beilishi Road, Xicheng District, Beijing 100037, China
| | - Xin-Hai Ni
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beilishi Road, Xicheng District, Beijing 100037, China
| | - Zhi-Hui Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beilishi Road, Xicheng District, Beijing 100037, China
| | - Jue Ye
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beilishi Road, Xicheng District, Beijing 100037, China
| | - Xian-Min Meng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beilishi Road, Xicheng District, Beijing 100037, China
| | - Zhi-Hong Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beilishi Road, Xicheng District, Beijing 100037, China
| | - Chang-Ming Xiong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beilishi Road, Xicheng District, Beijing 100037, China.
| | - Jian-Guo He
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College and Chinese Academy of Medical Sciences, Beilishi Road, Xicheng District, Beijing 100037, China.
| |
Collapse
|
28
|
Dolenc J, Šebeštjen M, Vrtovec B, Koželj M, Haddad F. Pulmonary hypertension in patients with advanced heart failure is associated with increased levels of interleukin-6. Biomarkers 2014; 19:385-90. [PMID: 24831174 DOI: 10.3109/1354750x.2014.918654] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Inflammatory, endothelial and neurohormonal biomarkers are involved in heart failure (HF) and pulmonary hypertension (PH) pathogenesis. OBJECTIVE To study these biomarkers in PH due to advanced HF. MATERIALS AND METHODS Thirty adults with HF were included. Interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), high-sensitivity C-reactive protein (hsCRP), endothelin-1 and N-terminal prohormone of brain natriuretic peptide (NT-proBNP) were measured in peripheral vein and pulmonary artery during right heart catheterisation. RESULTS IL-6, TNF-α, hsCRP and NT-proBNP correlated with pulmonary pressures independent of ventricular function, HF etiology and vascular bed. IL-6 was independent predictor of systolic pulmonary artery pressure (sPAP). DISCUSSION AND CONCLUSION Inflammatory biomarkers correlate to PH severity. IL-6 predicts sPAP in advanced HF.
Collapse
Affiliation(s)
- Jure Dolenc
- Department of Cardiology, University Medical Centre Ljubljana , Ljubljana , Slovenia
| | | | | | | | | |
Collapse
|
29
|
Vonk-Noordegraaf A, Haddad F, Chin KM, Forfia PR, Kawut SM, Lumens J, Naeije R, Newman J, Oudiz RJ, Provencher S, Torbicki A, Voelkel NF, Hassoun PM. Right heart adaptation to pulmonary arterial hypertension: physiology and pathobiology. J Am Coll Cardiol 2014; 62:D22-33. [PMID: 24355638 DOI: 10.1016/j.jacc.2013.10.027] [Citation(s) in RCA: 711] [Impact Index Per Article: 64.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 10/22/2013] [Indexed: 12/22/2022]
Abstract
Survival in patients with pulmonary arterial hypertension (PAH) is closely related to right ventricular (RV) function. Although pulmonary load is an important determinant of RV systolic function in PAH, there remains a significant variability in RV adaptation to pulmonary hypertension. In this report, the authors discuss the emerging concepts of right heart pathobiology in PAH. More specifically, the discussion focuses on the following questions. 1) How is right heart failure syndrome best defined? 2) What are the underlying molecular mechanisms of the failing right ventricle in PAH? 3) How are RV contractility and function and their prognostic implications best assessed? 4) What is the role of targeted RV therapy? Throughout the report, the authors highlight differences between right and left heart failure and outline key areas of future investigation.
Collapse
Affiliation(s)
| | - François Haddad
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, California
| | - Kelly M Chin
- Department of Internal Medicine, Pulmonary Division, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Paul R Forfia
- Pulmonary Hypertension and Right Heart Failure Program, Temple University Hospital, Philadelphia, Pennsylvania
| | - Steven M Kawut
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joost Lumens
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Robert Naeije
- Department of Pathophysiology, Faculty of Medicine, Free University of Brussels, Brussels, Belgium
| | - John Newman
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Ronald J Oudiz
- The David Geffen School of Medicine at UCLA, Liu Center for Pulmonary Hypertension, Division of Cardiology, Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California
| | - Steve Provencher
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Chemin Sainte-Foy, Québec, Canada
| | - Adam Torbicki
- Department of Pulmonary Circulation and Thromboembolic Diseases, Centre of Postgraduate Medical Education, ECZ, Otwock, Poland
| | - Norbert F Voelkel
- Division of Pulmonary and Critical Care Medicine and Victoria Johnson Lab for Lung Research, Virginia Commonwealth University, Richmond, Virginia; Johns Hopkins University, Baltimore, Maryland
| | - Paul M Hassoun
- Department of Internal Medicine, Pulmonary Division, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
30
|
Gu S, Su P, Yan J, Zhang X, An X, Gao J, Xin R, Liu Y. Comparison of gene expression profiles and related pathways in chronic thromboembolic pulmonary hypertension. Int J Mol Med 2013; 33:277-300. [PMID: 24337368 PMCID: PMC3896458 DOI: 10.3892/ijmm.2013.1582] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 12/03/2013] [Indexed: 01/08/2023] Open
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is one of the main causes of severe pulmonary hypertension. However, despite treatment (pulmonary endarterectomy), in approximately 15–20% of patients, pulmonary vascular resistance and pulmonary arterial pressure continue to increase. To date, little is known about the changes that occur in gene expression in CTEPH. The identification of genes associated with CTEPH may provide insight into the pathogenesis of CTEPH and may aid in diagnosis and treatment. In this study, we analyzed the gene expresion profiles of pulmonary artery endothelial cells from 5 patients with CTEPH and 5 healthy controls using oligonucleotide microarrays. Bioinformatics analyses using the Gene Ontology (GO) and KEGG databases were carried out to identify the genes and pathways specifically associated with CTEPH. Signal transduction networks were established to identify the core genes regulating the progression of CTEPH. A number of genes were found to be differentially expressed in the pulmonary artery endothelial cells from patients with CTEPH. In total, 412 GO terms and 113 pathways were found to be associated with our list of genes. All differential gene interactions in the Signal-Net network were analyzed. JAK3, GNA15, MAPK13, ARRB2 and F2R were the most significantly altered. Bioinformatics analysis may help gather and analyze large amounts of data in microarrays by means of rigorous experimental planning, scientific statistical analysis and the collection of complete data. In this study, a novel differential gene expression pattern was constructed. However, further studies are required to identify novel targets for the diagnosis and treatment of CTEPH.
Collapse
Affiliation(s)
- Song Gu
- Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Pixiong Su
- Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Jun Yan
- Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Xitao Zhang
- Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Xiangguang An
- Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Jie Gao
- Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Rui Xin
- Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Yan Liu
- Department of Cardiac Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| |
Collapse
|
31
|
Liu M, Ma Z, Guo X, Chen X, Yang Y, Wang C. Cardiovascular parameters of computed tomographic pulmonary angiography to assess pulmonary vascular resistance in patients with chronic thromboembolic pulmonary hypertension. Int J Cardiol 2013; 164:295-300. [DOI: 10.1016/j.ijcard.2011.07.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 06/18/2011] [Accepted: 07/03/2011] [Indexed: 10/17/2022]
|
32
|
Hagan G, Southwood M, Treacy C, Ross RM, Soon E, Coulson J, Sheares K, Screaton N, Pepke-Zaba J, Morrell NW, Rudd JHF. (18)FDG PET imaging can quantify increased cellular metabolism in pulmonary arterial hypertension: A proof-of-principle study. Pulm Circ 2012; 1:448-55. [PMID: 22530099 PMCID: PMC3329074 DOI: 10.4103/2045-8932.93543] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The past decade has seen increased application of 18-flurodeoxyglucose positron emission tomography (18FDG-PET) imaging to help diagnose and monitor disease, particularly in oncology, vasculitis and atherosclerosis. Disordered glycolytic metabolism and infiltration of plexiform lesions by inflammatory cells has been described in idiopathic pulmonary arterial hypertension (IPAH). We hypothesized that increased 18FDG uptake may be present in the lungs, large pulmonary arteries and right ventricle of patients with pulmonary hypertension, and that this uptake would be related to markers of immune activation. We imaged the thorax of 14 patients with pulmonary hypertension (idiopathic and chronic thromboembolic) and six controls by 18FDG-PET/computed tomography (CT) and measured uptake in the lung parenchyma, large pulmonary arteries and right ventricle. 18FDG uptake in the lungs and pulmonary arteries was normalized for venous blood activity to give a target-to-background ratio (TBR). Blood was contemporaneously drawn for high-sensitivity CRP - C-reactive protein (CRP) (hsCRP), N-Terminal Probrain natriuteric peptide (NT-ProBNP) and other inflammatory cytokines. IPAH patients had significantly higher lung parenchymal TBR (P=0.034) and right ventricle FDG uptake (P=0.007) than controls. Uptake in the main pulmonary arteries was similar in chronic thromboembolic pulmonary hypertension, IPAH and controls. There were no correlations between 18FDG uptake and hsCRP or inflammatory cytokine levels. NT-ProBNP correlated with RV uptake in those with pulmonary hypertension (r=0.55, P=0.04). In this pilot study, we found increased 18FDG uptake in the lung parenchyma and right ventricle of subjects with IPAH. The lung uptake might be useful as a surrogate marker of increased cellular metabolism and immune activation as underlying mechanisms in this disease. Further evaluation of the impact of targeted therapies in treatment-naïve patients and the significance of right ventricular uptake is suggested.
Collapse
Affiliation(s)
- Guy Hagan
- Pulmonary Vascular Disease Unit, Papworth Hospital, Papworth Everard, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Current world literature. Curr Opin Cardiol 2011; 26:356-61. [PMID: 21654380 DOI: 10.1097/hco.0b013e328348da50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
34
|
Larsen KO, Yndestad A, Sjaastad I, Løberg EM, Goverud IL, Halvorsen B, Jia J, Andreassen AK, Husberg C, Jonasson S, Lipp M, Christensen G, Aukrust P, Skjønsberg OH. Lack of CCR7 induces pulmonary hypertension involving perivascular leukocyte infiltration and inflammation. Am J Physiol Lung Cell Mol Physiol 2011; 301:L50-9. [PMID: 21498626 DOI: 10.1152/ajplung.00048.2010] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The chemokine receptor CCR7 regulates lymphocyte trafficking, and CCR7 deficiency induces infiltration of T and B cells adjacent to vessels in mouse lungs. Perivascular infiltration of T and B cells has also been found in human pulmonary arterial hypertension, and downregulation of the CCR7 receptor in circulating leukocytes of such patients has been observed. To investigate whether changes in the CCR7 system contribute to the pathogenesis of pulmonary hypertension, we utilized mice deficient of the CCR7 receptor. The cardiopulmonary and inflammatory responses of CCR7 depletion were evaluated in CCR7-deficient and wild-type mice. Measurements of cytokines upregulated in the animal model were also performed in patients with pulmonary hypertension and controls and in vascular smooth muscle cells. We found that mice lacking CCR7 had increased right ventricular systolic pressure, reduced pulmonary artery acceleration time, increased right ventricular/tibial length ratio, Rho kinase-mediated pulmonary vasoconstriction, and increased muscularization of distal arteries, indicating pulmonary hypertension. These mice also showed increased perivascular infiltration of leukocytes, consisting mainly of T and B cells, and increased mRNA levels of the inflammatory cytokines interleukin-12 and CX3CL1 within pulmonary tissue. Increased serum levels of interleukin-12 and CX3CL1 were also observed in patients with pulmonary hypertension, particularly in those with pulmonary hypertension associated with connective tissue disorder. In smooth muscle cells, interleukin-12 induced secretion of the angiogenic cytokine interleukin-8. We conclude that these results suggest a role for CCR7 in the development of pulmonary arterial hypertension, at least in some subgroups, possibly via pulmonary infiltration of lymphocytes and secretion of interleukin-12 and CX3CL1.
Collapse
Affiliation(s)
- Karl-Otto Larsen
- Department of Pulmonary Medicine, Oslo University Hospital Ullevål, Oslo, Norway.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Jaff MR, McMurtry MS, Archer SL, Cushman M, Goldenberg N, Goldhaber SZ, Jenkins JS, Kline JA, Michaels AD, Thistlethwaite P, Vedantham S, White RJ, Zierler BK. Management of massive and submassive pulmonary embolism, iliofemoral deep vein thrombosis, and chronic thromboembolic pulmonary hypertension: a scientific statement from the American Heart Association. Circulation 2011; 123:1788-830. [PMID: 21422387 DOI: 10.1161/cir.0b013e318214914f] [Citation(s) in RCA: 1551] [Impact Index Per Article: 110.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Venous thromboembolism (VTE) is responsible for the hospitalization of >250 000 Americans annually and represents a significant risk for morbidity and mortality. Despite the publication of evidence-based clinical practice guidelines to aid in the management of VTE in its acute and chronic forms, the clinician is frequently confronted with manifestations of VTE for which data are sparse and optimal management is unclear. In particular, the optimal use of advanced therapies for acute VTE, including thrombolysis and catheter-based therapies, remains uncertain. This report addresses the management of massive and submassive pulmonary embolism (PE), iliofemoral deep vein thrombosis (IFDVT),and chronic thromboembolic pulmonary hypertension (CTEPH). The goal is to provide practical advice to enable the busy clinician to optimize the management of patients with these severe manifestations of VTE. Although this document makes recommendations for management, optimal medical decisions must incorporate other factors, including patient wishes, quality of life, and life expectancy based on age and comorbidities. The appropriateness of these recommendations for a specific patient may vary depending on these factors and will be best judged by the bedside clinician.
Collapse
|