1
|
Shen HC, Lee WJ, Sun CY, Yu WK, Chen WC, Hsiao FY, Yang KY, Chen LK. Follistatin-respiratory connection predicting all-cause mortality among community-dwelling middle-to-old age individuals: Results from the I-Lan Longitudinal Study. J Nutr Health Aging 2024; 28:100285. [PMID: 38861881 DOI: 10.1016/j.jnha.2024.100285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/13/2024]
Abstract
OBJECTIVES The link between aging and pulmonary function decline is well-established, but the underlying mechanisms have yet to be fully revealed. Serum follistatin, a myokine implicated in muscle degeneration, may play a role in age-related pulmonary changes. This study aims to investigate the relationship between serum follistatin levels and pulmonary function decline in community-dwelling older adults, and evaluate their combined association with all-cause mortality. RESEARCH DESIGN AND METHODS This longitudinal cohort study utilized data from 751 participants aged ≥50 years in the I-Lan Longitudinal Aging Study between 2018-2019. Serum follistatin levels, spirometry results, demographic and clinical data were retrieved. Participants were stratified based on their follistatin levels. Survival curves and group comparisons based on follistatin levels and decline in peak expiratory flow (PEF) using Kaplan-Meier analysis and log-rank tests. Multivariate Cox proportional hazards models were further used to identify independent predictors of all-cause mortality during the 52-month follow-up. RESULTS Elevated follistatin levels significantly correlated with worse pulmonary function, particularly decreased PEF (p = 0.030). Kaplan-Meier analysis revealed the combination of elevated follistatin levels and decreased PEF was associated with increased risk of all-cause mortality (Log-rank p = 0.023). Cox proportional hazards models further identified that concurrent presence of higher follistatin levels and decreased PEF predicted higher risk of all-cause mortality (adjusted HR 3.58, 95% CI: 1.22-10.53, p = 0.020). CONCLUSION Higher serum follistatin levels correlate with decreased pulmonary function, specifically PEF decline, in community-dwelling older adults. Furthermore, the coexistence of elevated follistatin levels and decreased PEF was associated with risk of all-cause mortality. Follistatin may serve as a biomarker for pulmonary aging and related adverse outcomes.
Collapse
Affiliation(s)
- Hsiao-Chin Shen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Evidence-based Medicine, Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wei-Ju Lee
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Family Medicine, Taipei Veterans General Hospital Yuanshan Branch, Yilan, Taiwan
| | - Chuan-Yen Sun
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wen-Kuang Yu
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Chih Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Fei-Yuan Hsiao
- Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan; School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuang-Yao Yang
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Liang-Kung Chen
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan; Center for Geriatrics and Gerontology, Taipei Veterans General Hospital, Taipei, Taiwan; Taipei Municipal Gan-Dau Hospital (Managed by Taipei Veterans General Hospital), Taipei, Taiwan
| |
Collapse
|
2
|
Daoud A, Lema DA, Won T, Čiháková D. Integrative single-cell analysis of cardiac and pulmonary sarcoidosis using publicly available cardiac and bronchoalveolar lavage fluid sequencing datasets. Front Cardiovasc Med 2023; 10:1227818. [PMID: 37576111 PMCID: PMC10419306 DOI: 10.3389/fcvm.2023.1227818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/03/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Cardiac presentation of autoimmune sarcoidosis, known as cardiac sarcoidosis (CS), is a poorly understood disease with high mortality and low diagnosis rate. While CS is an immunological syndrome, little is known about how cardiac parenchymal and stromal cells mediate its pathogenesis. Moreover, while most current sarcoidosis research is based on research in pulmonary sarcoidosis (PS), it remains unclear how much both presentations of sarcoidosis overlap. To tackle these concerns, we leveraged publicly available sarcoidosis transcriptomic datasets. Methods Two publicly available bronchoalveolar lavage single-cell RNA sequencing datasets were integrated to analyze PS relative to control. Additionally, two publicly available cardiac single-nucleus RNA sequencing datasets were integrated to analyze CS relative to control. Following integration, we ran cell-cell communication, transcription factor, and differential expression analyses on parenchymal, stromal, and immune subsets identified in our analysis. Results Our analysis revealed that there was an expansion of stromal and immune cells in PS and CS. We also observed upregulation of Th17.1 and attenuated activation transcriptional profiles in the immune cells of CS and PS relative to control. Additionally, we found upregulation of pro-inflammatory and pro-fibrotic transcriptional profiles in the cardiac stromal cells of CS relative to control. We also found that cardiomyocytes exhibited upregulated cardiac stress and proliferation transcriptional profiles in CS relative to control. Conclusions Our integrative transcriptomic analysis shows that despite tissue-specific differences, there are shared transcriptional trends between CS and PS. It also shows that stromal and parenchymal populations exhibit transcriptional trends that could explain their pathogenic role in CS.
Collapse
Affiliation(s)
- Abdel Daoud
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Diego A. Lema
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Taejoon Won
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Daniela Čiháková
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
3
|
Fließer E, Lins T, Berg JL, Kolb M, Kwapiszewska G. The endothelium in lung fibrosis: a core signaling hub in disease pathogenesis? Am J Physiol Cell Physiol 2023; 325:C2-C16. [PMID: 37184232 DOI: 10.1152/ajpcell.00097.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/05/2023] [Accepted: 05/05/2023] [Indexed: 05/16/2023]
Abstract
Pulmonary fibrosis (PF) is a progressive chronic lung disease characterized by excessive deposition of extracellular matrix (ECM) and structural destruction, associated with a severe 5-year mortality rate. The onset of the disease is thought to be triggered by chronic damage to the alveolar epithelium. Since the pulmonary endothelium is an important component of the alveolar-capillary niche, it is also affected by the initial injury. In addition to ensuring proper gas exchange, the endothelium has critical functional properties, including regulation of vascular tone, inflammatory responses, coagulation, and maintenance of vascular homeostasis and integrity. Recent single-cell analyses have shown that shifts in endothelial cell (EC) subtypes occur in PF. Furthermore, the increased vascular remodeling associated with PF leads to deteriorated outcomes for patients, underscoring the importance of the vascular bed in PF. To date, the causes and consequences of endothelial and vascular involvement in lung fibrosis are poorly understood. Therefore, it is of great importance to investigate the involvement of EC and the vascular system in the pathogenesis of the disease. In this review, we will outline the current knowledge on the role of the pulmonary vasculature in PF, in terms of abnormal cellular interactions, hyperinflammation, vascular barrier disorders, and an altered basement membrane composition. Finally, we will summarize recent advances in extensive therapeutic research and discuss the significant value of novel therapies targeting the endothelium.
Collapse
Affiliation(s)
- Elisabeth Fließer
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Thomas Lins
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Johannes Lorenz Berg
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Martin Kolb
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Member of the German Lung Center (DZL), Cardiopulmonary Institute (CPI), Giessen, Germany
| |
Collapse
|
4
|
Wang Q, Xie Z, Wan N, Yang L, Jin Z, Jin F, Huang Z, Chen M, Wang H, Feng J. Potential biomarkers for diagnosis and disease evaluation of idiopathic pulmonary fibrosis. Chin Med J (Engl) 2023; 136:1278-1290. [PMID: 37130223 PMCID: PMC10309524 DOI: 10.1097/cm9.0000000000002171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Indexed: 05/04/2023] Open
Abstract
ABSTRACT Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease characterized by progressive lung fibrogenesis and histological features of usual interstitial pneumonia. IPF has a poor prognosis and presents a spectrum of disease courses ranging from slow evolving disease to rapid deterioration; thus, a differential diagnosis remains challenging. Several biomarkers have been identified to achieve a differential diagnosis; however, comprehensive reviews are lacking. This review summarizes over 100 biomarkers which can be divided into six categories according to their functions: differentially expressed biomarkers in the IPF compared to healthy controls; biomarkers distinguishing IPF from other types of interstitial lung disease; biomarkers differentiating acute exacerbation of IPF from stable disease; biomarkers predicting disease progression; biomarkers related to disease severity; and biomarkers related to treatment. Specimen used for the diagnosis of IPF included serum, bronchoalveolar lavage fluid, lung tissue, and sputum. IPF-specific biomarkers are of great clinical value for the differential diagnosis of IPF. Currently, the physiological measurements used to evaluate the occurrence of acute exacerbation, disease progression, and disease severity have limitations. Combining physiological measurements with biomarkers may increase the accuracy and sensitivity of diagnosis and disease evaluation of IPF. Most biomarkers described in this review are not routinely used in clinical practice. Future large-scale multicenter studies are required to design and validate suitable biomarker panels that have diagnostic utility for IPF.
Collapse
Affiliation(s)
- Qing Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Zhaoliang Xie
- Respiratory Department of Sanming Yong’an General Hospital, Sanming, Fujian 366000, China
| | - Nansheng Wan
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Lei Yang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhixian Jin
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Fang Jin
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhaoming Huang
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Min Chen
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Huiming Wang
- Department of Respiratory and Critical Care Medicine of Kunming Municipal First People's Hospital, Kunming, Yunnan 650000, China
| | - Jing Feng
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
5
|
Danila E, Aleksonienė R, Besusparis J, Gruslys V, Jurgauskienė L, Laurinavičienė A, Laurinavičius A, Mainelis A, Zablockis R, Zeleckienė I, Žurauskas E, Malickaitė R. Lymphocyte Subsets and Pulmonary Nodules to Predict the Progression of Sarcoidosis. Biomedicines 2023; 11:biomedicines11051437. [PMID: 37239108 DOI: 10.3390/biomedicines11051437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
The search for biological markers, which allow a relatively accurate assessment of the individual course of pulmonary sarcoidosis at the time of diagnosis, remains one of the research priorities in this field of pulmonary medicine. The aim of our study was to investigate possible prognostic factors for pulmonary sarcoidosis with a special focus on cellular immune inflammation markers. A 2-year follow-up of the study population after the initial prospective and simultaneous analysis of lymphocyte activation markers expression in the blood, as well as bronchoalveolar lavage fluid (BALF) and lung biopsy tissue of patients with newly diagnosed pulmonary sarcoidosis, was performed. We found that some blood and BAL fluid immunological markers and lung computed tomography (CT) patterns have been associated with a different course of sarcoidosis. We revealed five markers that had a significant negative association with the course of sarcoidosis (worsening pulmonary function tests and/or the chest CT changes)-blood CD4+CD31+ and CD4+CD44+ T lymphocytes, BALF CD8+CD31+ and CD8+CD103+ T lymphocytes and a number of lung nodules on chest CT at the time of the diagnosis. Cut-off values, sensitivity, specificity and odds ratio for predictors of sarcoidosis progression were calculated. These markers may be reasonable predictors of sarcoidosis progression.
Collapse
Affiliation(s)
- Edvardas Danila
- Clinic of Chest Diseases, Immunology and Allergology, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Regina Aleksonienė
- Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Justinas Besusparis
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, 08406 Vilnius, Lithuania
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Vygantas Gruslys
- Clinic of Chest Diseases, Immunology and Allergology, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Laimutė Jurgauskienė
- Clinic of Cardiac and Vascular Diseases, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Center of Laboratory Medicine, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Aida Laurinavičienė
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, 08406 Vilnius, Lithuania
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Arvydas Laurinavičius
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, 08406 Vilnius, Lithuania
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Antanas Mainelis
- Faculty of Mathematics and Informatics, Vilnius University, 03225 Vilnius, Lithuania
| | - Rolandas Zablockis
- Clinic of Chest Diseases, Immunology and Allergology, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Ingrida Zeleckienė
- Center of Radiology and Nuclear Medicine, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Edvardas Žurauskas
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, 08406 Vilnius, Lithuania
| | - Radvilė Malickaitė
- Clinic of Cardiac and Vascular Diseases, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Center of Laboratory Medicine, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| |
Collapse
|
6
|
Lu Y, Liu H, Dong B, Yang J, Kou L, Qin Q. Correlation between platelet-derived growth factor-B gene polymorphism and coronary heart disease. J Clin Lab Anal 2022; 36:e24683. [PMID: 36059119 PMCID: PMC9550974 DOI: 10.1002/jcla.24683] [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: 01/03/2022] [Revised: 06/26/2022] [Accepted: 08/14/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECT The aim of the present work was to investigate the correlation of plasma platelet-derived growth factor (PDGF)-BB level and single nucleotide polymorphism (SNP, rs1800817 and rs2285094) of PDGF-B gene with the onset and stability condition of coronary heart disease (CHD). METHODS Totally, 335 subjects were included in and divided into CHD (n = 247) and control group (n = 88) according to coronary angiography. Besides, the patients in the CHD group were divided into acute coronary syndrome (ACS) group (n = 165) and stable angina pectoria (SAP) group (n = 82), based on CHD stability condition. The plasma PDGF-BB level was measured by ELISA, and the genotype of PDGF-B was examined through qPCR assay. RESULTS The PDGF-BB level was positively correlated with hsCRP level (r = 0.149, p < 0.05). The genotype frequencies of SNP rs1800817 and rs2285094 match Hardy-Weinberg equilibrium. There was weak linkage disequilibrium between SNP rs1800817 and rs2285094: D' = 0.419, r2 = 0.04, which has no correlation with CHD. There was no statistical difference in plasma PDGF-BB level among different genotypes in rs1800817 and rs2285094. There were no differences in the plasma PDGF-BB level among patients with any genotype of SNP rs1800817 and rs2285094, no matter how it was grouped. Logistic regression results indicated that the plasma PDGF-BB level was the independent risk factor of CHD onset (OR = 1.003, 95% CI 1.001-1.006, p = 0.014). CONCLUSIONS High plasma PDGF-BB level is the risk factor of CHD and has correlation with instability of CHD. The plasma PDGF-BB level change may be related to inflammatory response. PDGF-B gene rs1800817 and rs2285094 polymorphisms are not correlated with CHD.
Collapse
Affiliation(s)
- Yaru Lu
- Department of Cardiology, Tianjin Chest Hospital, Chest Hospital Tianjin University, Tianjin, China
| | - Hui Liu
- Department of Hematology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Bo Dong
- Department of Cardiology, Tianjin Chest Hospital, Chest Hospital Tianjin University, Tianjin, China
| | - Jingyu Yang
- Department of Cardiology, Tianjin Chest Hospital, Chest Hospital Tianjin University, Tianjin, China
| | - Lu Kou
- Department of Cardiology, Tianjin Chest Hospital, Chest Hospital Tianjin University, Tianjin, China
| | - Qin Qin
- Department of Cardiology, Tianjin Chest Hospital, Chest Hospital Tianjin University, Tianjin, China
| |
Collapse
|
7
|
Byg KE, Illes Z, Sejbaek T, Lambertsen KL, Ellingsen T, Nielsen HH. Inflammatory profiles in plasma and cerebrospinal fluid of patients with neurosarcoidosis. J Neuroimmunol 2022; 367:577849. [DOI: 10.1016/j.jneuroim.2022.577849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/04/2022] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
|
8
|
Jahandideh A, Ståhle M, Virta J, Li XG, Liljenbäck H, Moisio O, Knuuti J, Roivainen A, Saraste A. Evaluation of [ 68Ga]Ga-NODAGA-RGD for PET Imaging of Rat Autoimmune Myocarditis. Front Med (Lausanne) 2022; 8:783596. [PMID: 34977085 PMCID: PMC8714834 DOI: 10.3389/fmed.2021.783596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/15/2021] [Indexed: 12/26/2022] Open
Abstract
The 68Gallium-labeled 1,4,7-triazacyclononane-1-glutaric acid-4,7-diacetic acid conjugated radiolabelled arginine-glycine-aspartic acid peptide ([68Ga]Ga-NODAGA-RGD) is a positron emission tomography (PET) tracer binding to cell surface receptor αvβ3 integrin that is upregulated during angiogenesis and inflammation. We studied whether αvβ3 targeting PET imaging can detect myocardial inflammation in a rat model of autoimmune myocarditis. To induce myocarditis, rats (n = 8) were immunized with porcine cardiac myosin in complete Freund's adjuvant on days 0 and 7. Control rats (n = 8) received Freund's adjuvant alone. On day 21, in vivo PET/CT imaging with [68Ga]Ga-NODAGA-RGD followed by ex vivo autoradiography and immunohistochemistry were carried out. Inflammatory lesions were detected histologically in the myocardium of 7 out of 8 immunized rats. In vivo PET images showed higher [68Ga]Ga-NODAGA-RGD accumulation in the myocardium of rats with inflammation than the non-inflamed myocardium of control rats (SUVmean 0.4 ± 0.1 vs. 0.1 ± 0.02; P = 0.00006). Ex vivo autoradiography and histology confirmed that [68Ga]Ga-NODAGA-RGD uptake co-localized with inflammatory lesions containing αvβ3 integrin-positive capillary-like structures. A non-specific [68Ga]Ga-DOTA-(RGE)2 tracer showed 76% lower uptake than [68Ga]Ga-NODAGA-RGD in the inflamed myocardium. Our results indicate that αvβ3 integrin-targeting [68Ga]Ga-NODAGA-RGD is a potential PET tracer for the specific detection of active inflammatory lesions in autoimmune myocarditis.
Collapse
Affiliation(s)
| | - Mia Ståhle
- Turku PET Centre, University of Turku, Turku, Finland
| | - Jenni Virta
- Turku PET Centre, University of Turku, Turku, Finland
| | - Xiang-Guo Li
- Turku PET Centre, University of Turku, Turku, Finland
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Olli Moisio
- Turku PET Centre, University of Turku, Turku, Finland
| | - Juhani Knuuti
- Turku PET Centre, University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Antti Saraste
- Turku PET Centre, University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland.,Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
9
|
Yokoyama A, Sakamoto Y, Jo T, Urushiyama H, Tamiya H, Tanaka G, Matsui H, Fushimi K, Yasunaga H, Nagase T. Pulmonary disease as a risk factor for transfusion-related acute lung injury. ERJ Open Res 2021; 7:00039-2021. [PMID: 34476252 PMCID: PMC8405876 DOI: 10.1183/23120541.00039-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/20/2021] [Indexed: 12/03/2022] Open
Abstract
Transfusion-related acute lung injury (TRALI) is a severe condition characterised by noncardiogenic pulmonary oedema that develops within 6 h of blood transfusion. Patient factors and blood products have both been implicated in the development of TRALI; however, the role of pulmonary disease has not been investigated. We aimed to determine whether pulmonary disease is a risk factor for TRALI. We conducted a nested case-control study using data from the Diagnosis Procedure Combination database, a nationwide inpatient database in Japan, between July 2010 and March 2015. Case patients who developed TRALI were 1:4-matched with control patients for sex, age and same hospital for receipt of blood transfusion. We conducted a multivariable conditional logistic regression analysis to evaluate the associations of TRALI with various factors including comorbidities, body mass index (BMI) and plasma-containing blood products. We identified 2 019 501 hospitalised patients who received a blood transfusion. Among these patients, 72 developed TRALI. The 72 case patients had higher proportions of haematological malignancy, trauma and interstitial lung disease (ILD) than the 288 matched control patients. The multivariable conditional logistic regression analysis showed that occurrence of TRALI was associated with ILD (odds ratio, 3.88; 95% confidence interval, 1.11–13.6), BMI ≥25.0 kg·m−2 (2.10; 1.05–4.24) and plasma-containing blood products (1.94; 1.10–3.42), but not with infectious lung disease or obstructive airway disease. In conclusion, ILD was an independent risk factor for the development of TRALI. Physicians should be aware of the increased risk of TRALI in patients with ILD. Physicians should be aware of the risk of developing transfusion-related acute lung injury when performing a blood transfusion in patients with interstitial lung disease.https://bit.ly/3jdoZQP
Collapse
Affiliation(s)
- Akira Yokoyama
- Dept of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukiyo Sakamoto
- Dept of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taisuke Jo
- Dept of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Dept of Health Services Research, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hirokazu Urushiyama
- Dept of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Tamiya
- Dept of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Goh Tanaka
- Dept of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroki Matsui
- Dept of Clinical Epidemiology and Health Economics, School of Public Health, The University of Tokyo, Tokyo, Japan
| | - Kiyohide Fushimi
- Dept of Health Policy and Informatics, Tokyo Medical and Dental University Graduate School of Medicine, Tokyo, Japan
| | - Hideo Yasunaga
- Dept of Clinical Epidemiology and Health Economics, School of Public Health, The University of Tokyo, Tokyo, Japan
| | - Takahide Nagase
- Dept of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
10
|
Hirani N, MacKinnon AC, Nicol L, Ford P, Schambye H, Pedersen A, Nilsson UJ, Leffler H, Sethi T, Tantawi S, Gravelle L, Slack RJ, Mills R, Karmakar U, Humphries D, Zetterberg F, Keeling L, Paul L, Molyneaux PL, Li F, Funston W, Forrest IA, Simpson AJ, Gibbons MA, Maher TM. Target inhibition of galectin-3 by inhaled TD139 in patients with idiopathic pulmonary fibrosis. Eur Respir J 2021; 57:13993003.02559-2020. [PMID: 33214209 PMCID: PMC8156151 DOI: 10.1183/13993003.02559-2020] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022]
Abstract
Galectin (Gal)-3 is a profibrotic β-galactoside-binding lectin that plays a key role in the pathogenesis of idiopathic pulmonary fibrosis (IPF) and IPF exacerbations. TD139 is a novel and potent small-molecule inhibitor of Gal-3. A randomised, double-blind, multicentre, placebo-controlled, phase 1/2a study was conducted to assess the safety, tolerability, pharmacokinetics and pharmacodynamics of inhaled TD139 in 36 healthy subjects and 24 patients with IPF. Six dose cohorts of six healthy subjects were evaluated (4:2 TD139:placebo ratio) with single doses of TD139 (0.15–50 mg) and three dose cohorts of eight patients with IPF (5:3 TD139:placebo ratio) with once-daily doses of TD139 (0.3–10 mg) for 14 days. Inhaled TD139 was well tolerated with no significant treatment-related side-effects. TD139 was rapidly absorbed, with mean time taken to reach maximum plasma concentration (Cmax) values ranging from 0.6 to 3 h and a plasma half-life (T1/2) of 8 h. The concentration of TD139 in the lung was >567-fold higher than in the blood, with systemic exposure predicting exposure in the target compartment. Gal-3 expression on alveolar macrophages was reduced in the 3 and 10 mg dose groups compared with placebo, with a concentration-dependent inhibition demonstrated. Inhibition of Gal-3 expression in the lung was associated with reductions in plasma biomarkers centrally relevant to IPF pathobiology (platelet-derived growth factor-BB, plasminogen activator inhibitor-1, Gal-3, CCL18 and YKL-40). TD139 is safe and well tolerated in healthy subjects and IPF patients. It was shown to suppress Gal-3 expression on bronchoalveolar lavage macrophages and, in a concerted fashion, decrease plasma biomarkers associated with IPF progression. TD139 is a potent inhibitor of galectin-3, a key driver of fibrosis in the lung. In this phase 1/2a clinical study, inhaled TD139 was safe, well tolerated, and demonstrated target engagement and decreased plasma biomarkers associated with IPF progression.https://bit.ly/2JREKx6
Collapse
Affiliation(s)
- Nikhil Hirani
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Alison C MacKinnon
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.,Galecto, Copenhagen, Denmark
| | - Lisa Nicol
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | | | | | | | | | - Hakon Leffler
- Dept of Laboratory Medicine, Lund University, Lund, Sweden
| | | | | | | | | | - Ross Mills
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Utsa Karmakar
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Duncan Humphries
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | | | | | - Lyn Paul
- National Institute for Health Research Respiratory Clinical Research Facility, Royal Brompton and Harefield NHS Foundation Trust, and Fibrosis Research Group, National Heart and Lung Institute, Imperial College London, London, UK
| | - Philip L Molyneaux
- National Institute for Health Research Respiratory Clinical Research Facility, Royal Brompton and Harefield NHS Foundation Trust, and Fibrosis Research Group, National Heart and Lung Institute, Imperial College London, London, UK
| | - Feng Li
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Wendy Funston
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Ian A Forrest
- Respiratory Medicine Unit, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - A John Simpson
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.,Respiratory Medicine Unit, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Michael A Gibbons
- Respiratory Dept, Institute of Biomedical and Clinical Science, Royal Devon and Exeter NHS Foundation Trust, Medical School, University of Exeter, Exeter, UK
| | - Toby M Maher
- National Institute for Health Research Respiratory Clinical Research Facility, Royal Brompton and Harefield NHS Foundation Trust, and Fibrosis Research Group, National Heart and Lung Institute, Imperial College London, London, UK.,Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
11
|
Aleksonienė R, Besusparis J, Gruslys V, Jurgauskienė L, Laurinavičienė A, Laurinavičius A, Malickaitė R, Norkūnienė J, Zablockis R, Žurauskas E, Danila E. CD31 +, CD38 +, CD44 +, and CD103 + lymphocytes in peripheral blood, bronchoalveolar lavage fluid and lung biopsy tissue in sarcoid patients and controls. J Thorac Dis 2021; 13:2300-2318. [PMID: 34012580 PMCID: PMC8107533 DOI: 10.21037/jtd-20-2396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background The mechanisms driving the transition from inflammation to fibrosis in sarcoidosis patients are poorly understood; prognostic features are lacking. Immune cell profiling may provide insights into pathogenesis and prognostic factors of the disease. This study aimed to establish associations in simultaneous of lymphocyte subset profiles in the blood, bronchoalveolar lavage fluid (BALF), and lung biopsy tissue in the patients with newly diagnosed sarcoidosis. Methods A total of 71 sarcoid patients (SPs) and 20 healthy controls (HCs) were enrolled into the study. CD31, CD38, CD44, CD103 positive T lymphocytes in blood and BALF were analysed. Additionally, the densities of CD4, CD8, CD38, CD44, CD103 positive cells in lung tissue biopsies were estimated by digital image analysis. Results Main findings: (I) increase of percentage of CD3+CD4+CD38+ in BALF and blood, and increase of percentage of CD3+CD4+CD44+ in BALF in Löfgren syndrome patients comparing with patients without Löfgren syndrome, (II) increase of percentage of CD3+CD4+103+ in BALF and in blood in patients without Löfgren syndrome (comparing with Löfgren syndrome patients) and increase of percentage of CD3+CD4+103+ in BALF and in blood in more advanced sarcoidosis stage. (III) Increasing percentage of BALF CD3+CD4+CD31+ in sarcoidosis patients when comparing with controls independently of presence of Löfgren syndrome, smoking status or stage of sarcoidosis. Several significant correlations were found. Conclusions Lymphocyte subpopulations in blood, BALF, and lung tissue were substantially different in SPs at the time of diagnosis compared to HCs. CD3+CD4+CD31+ in BALF might be a potential supporting marker for the diagnosis of sarcoidosis. CD3+CD4+CD38+ in BALF and blood and CD3+CD4+CD44+ in BALF may be markers of the acute immune response in sarcoidosis patients. CD4+CD103+ T-cells in BALF and in blood are markers of the persistent immune response in sarcoidosis patients and are potential prognostic features of the chronic course of this disease.
Collapse
Affiliation(s)
- Regina Aleksonienė
- Clinic of Chest Diseases, Immunology and Allergology of Faculty of Medicine, Vilnius University, Vilnius, Lithuania.,Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Justinas Besusparis
- National Center of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania.,Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Vygantas Gruslys
- Clinic of Chest Diseases, Immunology and Allergology of Faculty of Medicine, Vilnius University, Vilnius, Lithuania.,Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | | | - Aida Laurinavičienė
- National Center of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania.,Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Arvydas Laurinavičius
- National Center of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania.,Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | | | - Jolita Norkūnienė
- Department of Mathematical Statistics, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Rolandas Zablockis
- Clinic of Chest Diseases, Immunology and Allergology of Faculty of Medicine, Vilnius University, Vilnius, Lithuania.,Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Edvardas Žurauskas
- National Center of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania.,Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Edvardas Danila
- Clinic of Chest Diseases, Immunology and Allergology of Faculty of Medicine, Vilnius University, Vilnius, Lithuania.,Center of Pulmonology and Allergology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| |
Collapse
|
12
|
Biener L, Kruse J, Tuleta I, Pizarro C, Kreuter M, Birring SS, Nickenig G, Skowasch D. Association of proangiogenic and profibrotic serum markers with lung function and quality of life in sarcoidosis. PLoS One 2021; 16:e0247197. [PMID: 33617593 PMCID: PMC7899331 DOI: 10.1371/journal.pone.0247197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 02/02/2021] [Indexed: 11/18/2022] Open
Abstract
Background Sarcoidosis is a systemic inflammatory granulomatous disease, frequently affecting the lung. If left untreated, it may end in lung fibrosis. Proangiogenic and profibrotic vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-β1, fibroblast growth factor (FGF)-2 and platelet-derived growth factor (PDGF)-AB are a known therapeutical target in pulmonary fibrosing diseases, e.g. IPF, but there is no targeted therapy option for pulmonary fibrosis in sarcoidosis. Objectives The aim of our study was to determine the association of these markers’ serum levels on lung function and the patients’ quality of life in a long-term follow-up of sarcoidosis patients, to provide further information for finding targeted therapy options for pulmonary sarcoidosis. Methods 54 patients with sarcoidosis underwent blood sampling, pulmonary function testing and answered the King’s Brief Interstitial Lung Disease (K-BILD) questionnaire at baseline and at three-years follow-up. Serum levels of profibrotic and angiogenic markers were assessed at baseline by enzyme-linked immunosorbent assay. Results Between 2015 and 2018, 54 patients with biopsy proven sarcoidosis were enrolled. Throughout the observation period, there was a significant decrease in the diffusion capacity for carbon monoxide (DLCO) [%] (-6.5504 ± 13,39, p = 0.001) and forced expiratory volume in one second predicted (FEV1) [%] (-6.07 ± 12.09, p = 0.001). Patients with greater impairment of forced vital capacity (FVC) did have significantly higher serum levels of VEGF (p = 0.03) and PDGF-AB (p<0.001). The K-BILD questionnaire did not change significantly during follow-up. However, patients with worsening K-BILD scores did have significantly higher serum-levels of PDGF-AB (2.67 pg/ml ± 0.93 vs. 1.88 pg/ml ± 0.60, p = 0.004) at baseline, compared to those with unchanged or increasing K-BILD scores. Conclusions Among patients with pulmonary sarcoidosis, baseline serum levels of VEGF and PDGF-AB were associated with pulmonary function impairment. Furthermore, PDGF-AB was associated with worsening K-BILD scores. No such association was observed for FGF-2 and TGF-ß1. VEGF and PDGF-AB may be possible prognostic and therapeutic targets in sarcoidosis as a fibrosing ILD beyond IPF.
Collapse
Affiliation(s)
- L. Biener
- Department of Internal Medicine II–Cardiology, Pneumology and Angiology, University Hospital Bonn, Bonn, Germany
- * E-mail:
| | - J. Kruse
- Department of Internal Medicine II–Cardiology, Pneumology and Angiology, University Hospital Bonn, Bonn, Germany
| | - I. Tuleta
- Department of Cardiology I, University Hospital Muenster, Muenster, Germany
| | - C. Pizarro
- Department of Internal Medicine II–Cardiology, Pneumology and Angiology, University Hospital Bonn, Bonn, Germany
| | - M. Kreuter
- Centre for Interstitial and Rare Lung Diseases, Pneumology, Thoraxklinik, University of Heidelberg, Germany and German Centre for Lung Research, Heidelberg, Germany
| | - S. S. Birring
- Centre for Human & Applied Physiological Sciences, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - G. Nickenig
- Department of Internal Medicine II–Cardiology, Pneumology and Angiology, University Hospital Bonn, Bonn, Germany
| | - D. Skowasch
- Department of Internal Medicine II–Cardiology, Pneumology and Angiology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
13
|
Exhaled Biomarkers in Idiopathic Pulmonary Fibrosis-A Six-Month Follow-Up Study in Patients Treated with Pirfenidone. J Clin Med 2020; 9:jcm9082523. [PMID: 32764328 PMCID: PMC7465603 DOI: 10.3390/jcm9082523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/11/2020] [Accepted: 08/03/2020] [Indexed: 12/17/2022] Open
Abstract
The mechanism of action of pirfenidone in idiopathic pulmonary fibrosis (IPF) has not been fully elucidated. To offer additional insight, we evaluated the change in the cytokine profile in exhaled breath condensate (EBC) following a six-month treatment with pirfenidone in patients with IPF. EBC concentrations of interleukin (IL)-6, IL-8, IL-15, TNF-α and VEGF-A were assessed with ELISA and compared at baseline and after six months of pirfenidone treatment. Twenty-nine patients with IPF and 13 controls were evaluated at baseline. With the exception of IL-8 concentration, which was lower in patients with IPF when compared to controls (p = 0.005), the cytokine levels did not differ between the groups. Despite the use of a high sensitivity assay, IL-8 reached detectable values only in 24% of IPF patients. EBC analysis after six months of treatment with pirfenidone did not reveal any differences in the cytokine levels. The change in EBC vascular endothelial growth factor A (VEGF-A) correlated with the change in the 6 min walk distance (r = 0.54, p = 0.045). We conclude that a six-month treatment with pirfenidone did not significantly change the EBC cytokine profile. Our findings support the potential usefulness of VEGF-A as a marker in IPF. The low EBC IL-8 level in patients with IPF is a novel finding which needs confirmation in larger studies.
Collapse
|
14
|
Ramos-Casals M, Retamozo S, Sisó-Almirall A, Pérez-Alvarez R, Pallarés L, Brito-Zerón P. Clinically-useful serum biomarkers for diagnosis and prognosis of sarcoidosis. Expert Rev Clin Immunol 2019; 15:391-405. [DOI: 10.1080/1744666x.2019.1568240] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Manuel Ramos-Casals
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- SarcoGEAS-SEMI Study Group, Study Group of Autoimmune Diseases (GEAS), Spanish Society of Internal Medicine (SEMI), Spain
| | - Soledad Retamozo
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- Rheumatology Unit, Instituto Modelo de Cardiología Privado S.R.L, Córdoba, Argentina
- Rheumatology Unit, Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Córdoba, Argentina
- Instituto De Investigaciones En Ciencias De La Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Antoni Sisó-Almirall
- Centre d’Assistència Primària ABS Les Corts, CAPSBE, Barcelona, Spain
- Primary Healthcare Transversal Research Group, IDIBAPS, Barcelona, Spain
| | - Roberto Pérez-Alvarez
- SarcoGEAS-SEMI Study Group, Study Group of Autoimmune Diseases (GEAS), Spanish Society of Internal Medicine (SEMI), Spain
- Department of Internal Medicine, Hospital Alvaro Cunqueiro, Vigo, Spain
| | - Lucio Pallarés
- SarcoGEAS-SEMI Study Group, Study Group of Autoimmune Diseases (GEAS), Spanish Society of Internal Medicine (SEMI), Spain
- Systemic Autoimmune Diseases Uni, Department of Internal Medicine, Hospital de Son Espases, Palma de Mallorca, Spain
| | - Pilar Brito-Zerón
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- SarcoGEAS-SEMI Study Group, Study Group of Autoimmune Diseases (GEAS), Spanish Society of Internal Medicine (SEMI), Spain
- Autoimmune Diseases Unit, Department of Medicine, Hospital CIMA, Sanitas, Spain
| |
Collapse
|
15
|
Proangiogenic and Profibrotic Markers in Pulmonary Sarcoidosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1114:57-66. [DOI: 10.1007/5584_2018_199] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
|
16
|
Bale S, Venkatesh P, Sunkoju M, Godugu C. An Adaptogen: Withaferin A Ameliorates in Vitro and in Vivo Pulmonary Fibrosis by Modulating the Interplay of Fibrotic, Matricelluar Proteins, and Cytokines. Front Pharmacol 2018; 9:248. [PMID: 29623041 PMCID: PMC5874319 DOI: 10.3389/fphar.2018.00248] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/06/2018] [Indexed: 12/16/2022] Open
Abstract
Pulmonary fibrosis (PF) is chronic lung disease with only two FDA approved clinically available drugs, with limited safety profile. Inadequate therapy motivated us to explore the effect of vimentin inhibitor Withaferin A, as an anti-fibrotic agent against TGF-β1-induced in vitro fibrotic events and Bleomycin induced in vivo fibrosis with an emphasis on epithelial to mesenchymal transition (EMT), extracellular matrix deposition (ECM), inflammation, and angiogenesis. In vitro EMT and fibrotic events were induced by TGF-β1 in alveolar epithelial cells and human fetal lung fibroblasts followed by treatment with Withaferin A (0.25, 0.5, and 1 μM concentrations) to explore its anti-fibrotic effects. In vivo potential of Withaferin A (2 and 4 mg/kg) was assessed in murine model of Bleomycin induced PF. All the parameters and molecular studies related to PF were performed at the end of treatment period. Withaferin A treatment reduced the progression of PF by modulating the EMT related cell markers both in vivo and in vitro. Withaferin A ameliorated the expression of inflammatory cytokines including NF-κB p65, IL-1β and TNF-α, as well as attenuated the expression of pro-fibrotic proteins including CTGF, collagen 1A2, collagen 3A1, and fibronectin. Expression of angiogenic factors like VEGF, FAK, p38 MAPK, and PLC-γ1 were also inhibited by Withaferin A. Phosphorylation of Smad 2/3 induced by TGF-β1 and Bleomycin were significantly inhibited. Withaferin A suppressed expression of pro-inflammatory, pro-fibrotic, and pro-angiogenic mediators and also reduced the ECM deposition. In a nutshell, Withaferin A could probably prove as an efficient and potential therapeutic against PF.
Collapse
Affiliation(s)
- Swarna Bale
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Pooladanda Venkatesh
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Manoj Sunkoju
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| |
Collapse
|
17
|
Treprostinil inhibits proliferation and extracellular matrix deposition by fibroblasts through cAMP activation. Sci Rep 2018; 8:1087. [PMID: 29348469 PMCID: PMC5773699 DOI: 10.1038/s41598-018-19294-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 10/09/2017] [Indexed: 12/25/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by peripheral lung fibrosis and increased interstitial extracellular matrix (ECM) deposition. In IPF, tumor growth factor (TGF)-β1 which is the major stimulus of ECM deposition, and platelet derived growth factor (PDGF)-BB is a potent stimulus of fibrosis. Thus, the effect of Treprostinil on TGF-ß1 and PDGF-induced fibroblast proliferation and ECM deposition was investigated. Human peripheral lung fibroblasts of seven IPF patients and five lung donors were stimulated by PDGF, or TGF-β1, or the combination. Cells were pre-incubated (30 min) with either Treprostinil, forskolin, di-deoxyadenosine (DDA), or vehicle. Treprostinil time dependently activated cAMP thereby preventing PDGF-BB induced proliferation and TGF-β1 secretion. Cell counts indicated proliferation; α-smooth muscle actin (α-SMA) indicted differentiation, and collagen type-1 or fibronectin deposition remodeling. Myo-fibroblast indicating α-SMA expression was significantly reduced and its formation was altered by Treprostinil. Collagen type-I and fibronectin deposition were also reduced by Treprostinil. The effect of Treprostinil on collagen type-I deposition was cAMP sensitive as it was counteracted by DDA, while the effect on fibronectin was not cAMP mediated. Treprostinil antagonized the pro-fibrotic effects of both PDGF-BB and TGF-β1 in primary human lung fibroblasts. The data presented propose a therapeutic relevant anti-fibrotic effect of Treprostinil in IPF.
Collapse
|
18
|
Yang L, Herrera J, Gilbertsen A, Xia H, Smith K, Benyumov A, Bitterman PB, Henke CA. IL-8 mediates idiopathic pulmonary fibrosis mesenchymal progenitor cell fibrogenicity. Am J Physiol Lung Cell Mol Physiol 2017; 314:L127-L136. [PMID: 28860143 PMCID: PMC5866425 DOI: 10.1152/ajplung.00200.2017] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease, but the mechanisms driving progression remain incompletely defined. We previously reported that the IPF lung harbors fibrogenic mesenchymal progenitor cells (MPCs), which serve as a cell of origin for IPF fibroblasts. Proliferating IPF MPCs are located at the periphery of fibroblastic foci in an active cellular front at the interface between the myofibroblast-rich focus core and adjacent normal alveolar structures. Among a large set of genes that distinguish IPF MPCs from their control counterparts, we identified IL-8 as a candidate mediator of IPF MPC fibrogenicity and driver of fibrotic progression. IPF MPCs and their progeny displayed increased steady-state levels of IL-8 and its cognate receptor CXCR1 and secreted more IL-8 than did controls. IL-8 functioned in an autocrine manner promoting IPF MPC self-renewal and the proliferation and motility of IPF MPC progeny. Secreted IL-8 also functioned in a paracrine manner stimulating macrophage migration. Analysis of IPF lung tissue demonstrated codistribution of IPF MPCs with activated macrophages in the active cellular front of the fibroblastic focus. These findings indicate that IPF MPC-derived IL-8 is capable of expanding the mesenchymal cell population and recruiting activated macrophages cells to actively evolving fibrotic lesions.
Collapse
Affiliation(s)
- Libang Yang
- Department of Medicine, University of Minnesota , Minneapolis, Minnesota
| | - Jeremy Herrera
- Department of Medicine, University of Minnesota , Minneapolis, Minnesota
| | - Adam Gilbertsen
- Department of Medicine, University of Minnesota , Minneapolis, Minnesota
| | - Hong Xia
- Department of Medicine, University of Minnesota , Minneapolis, Minnesota
| | - Karen Smith
- Department of Medicine, University of Minnesota , Minneapolis, Minnesota
| | - Alexey Benyumov
- Department of Medicine, University of Minnesota , Minneapolis, Minnesota
| | - Peter B Bitterman
- Department of Medicine, University of Minnesota , Minneapolis, Minnesota
| | - Craig A Henke
- Department of Medicine, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|
19
|
Li X, Yue S, Luo Z. Mesenchymal stem cells in idiopathic pulmonary fibrosis. Oncotarget 2017; 8:102600-102616. [PMID: 29254275 PMCID: PMC5731985 DOI: 10.18632/oncotarget.18126] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/07/2017] [Indexed: 12/21/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a major cause of respiratory failure in critically ill patients and common outcome of various lung interstitial diseases. Its mortality remains high, and no effective pharmacotherapy, in addition to artificial ventilation and transplantation, exists. As such, the administration of mesenchymal stem or stromal cells (MSCs) is currently investigated as a new therapeutic method for pulmonary fibrosis. Clinical trials on MSC-based therapy as a potential treatment for lung injury and fibrosis are also performed. MSCs can migrate to injured sites and secrete multiple paracrine factors and then regulate endothelial and epithelial permeability, decrease inflammation, enhance tissue repair, and inhibit bacterial growth. In this review, recent studies on stem cells, particularly MSCs, involved in alleviating lung inflammation and fibrosis and their potential MSC-induced mechanisms, including migration and differentiation, soluble factor and extracellular vesicle secretion, and endogenous regulatory functions, were summarized.
Collapse
Affiliation(s)
- Xiaohong Li
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Shaojie Yue
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
| | - Ziqiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
20
|
Alves MJ, Figuerêdo RG, Azevedo FF, Cavallaro DA, Neto NIP, Lima JDC, Matos-Neto E, Radloff K, Riccardi DM, Camargo RG, De Alcântara PSM, Otoch JP, Junior MLB, Seelaender M. Adipose tissue fibrosis in human cancer cachexia: the role of TGFβ pathway. BMC Cancer 2017; 17:190. [PMID: 28288584 PMCID: PMC5348844 DOI: 10.1186/s12885-017-3178-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/08/2017] [Indexed: 02/06/2023] Open
Abstract
Background Cancer cachexia is a multifactorial syndrome that dramatically decreases survival. Loss of white adipose tissue (WAT) is one of the key characteristics of cachexia. WAT wasting is paralleled by microarchitectural remodeling in cachectic cancer patients. Fibrosis results from uncontrolled ECM synthesis, a process in which, transforming growth factor-beta (TGFβ) plays a pivotal role. So far, the mechanisms involved in adipose tissue (AT) re-arrangement, and the role of TGFβ in inducing AT remodeling in weight-losing cancer patients are poorly understood. This study examined the modulation of ECM components mediated by TGFβ pathway in fibrotic AT obtained from cachectic gastrointestinal cancer patients. Methods After signing the informed consent form, patients were enrolled into the following groups: cancer cachexia (CC, n = 21), weight-stable cancer (WSC, n = 17), and control (n = 21). The total amount of collagen and elastic fibers in the subcutaneous AT was assessed by histological analysis and by immunohistochemistry. TGFβ isoforms expression was analyzed by Multiplex assay and by immunohistochemistry. Alpha-smooth muscle actin (αSMA), fibroblast-specific protein (FSP1), Smad3 and 4 were quantified by qPCR and/or by immunohistochemistry. Interleukin (IL) 2, IL5, IL8, IL13 and IL17 content, cytokines known to be associated with fibrosis, was measured by Multiplex assay. Results There was an accumulation of collagen and elastic fibers in the AT of CC, as compared with WSC and controls. Collagens type I, III, VI, and fibronectin expression was enhanced in the tissue of CC, compared with both WSC and control. The pronounced expression of αSMA in the surrounding of adipocytes, and the increased mRNA content for FSP1 (20-fold) indicate the presence of activated myofibroblasts; particularly in CC. TGFβ1 and TGFβ3 levels were up-regulated by cachexia in AT, as well in the isolated adipocytes. Smad3 and Smad4 labeling was found to be more evident in the fibrotic areas of CC adipose tissue. Conclusions Cancer cachexia promotes the development of AT fibrosis, in association with altered TGFβ signaling, compromising AT organization and function.
Collapse
Affiliation(s)
- Michele Joana Alves
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| | - Raquel Galvão Figuerêdo
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Diego Alexandre Cavallaro
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.,Department of Nutrition, Federal University of Sao Paulo, Sao Paulo, Brazil
| | | | - Joanna Darck Carola Lima
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Emidio Matos-Neto
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Katrin Radloff
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Daniela Mendes Riccardi
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Rodolfo Gonzalez Camargo
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | - José Pinhata Otoch
- Department of Surgery, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil.,Department of Clinical Surgery, Hospital University, University of Sao Paulo, Sao Paulo, Brazil
| | - Miguel Luiz Batista Junior
- Biotechnology Group, Laboratory of Adipose Tissue Biology, University of Mogi das Cruzes, Mogi das Cruzes, Brazil
| | - Marília Seelaender
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.,Department of Surgery, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|