1
|
Huang JX, Xiao BJ, Yan YX, Xie W, Feng LY, Liu XM. Association Between Visceral Adipose Tissue and Chronic Respiratory Diseases: A Two-Sample Multivariable Mendelian Randomization Study in European Population. Int J Chron Obstruct Pulmon Dis 2025; 20:919-928. [PMID: 40191268 PMCID: PMC11972585 DOI: 10.2147/copd.s510828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/22/2025] [Indexed: 04/09/2025] Open
Abstract
Background The relationship between obesity and some respiratory diseases has been well documented. However there have been few studies on the association between visceral adipose tissue (VAT) and chronic respiratory diseases (CRDs), it remains unclear whether VAT is causally associated with CRDs. Methods We used two-sample Mendelian randomization (MR) to illuminate the effects of VAT on four CRDs: chronic obstructive pulmonary disease (COPD), allergic asthma, interstitial lung disease (ILD), and sarcoidosis. Inverse variance weighted (IVW) served as the primary assessment method. MR Egger, weighted median, Simple mode and Weighted mode were the supplementary methods for MR analysis. We used multivariate MR analysis to adjust for the effect of body mass index (BMI) on outcomes, Egger intercept, MR-pleiotropy residual sum and outlier, and leave-one-out analysis to confirm the MR results' consistency. Results Genetically-predicted VAT was associated with an increased risk of COPD (OR = 1.56; 95% CI: 1.34-1.82; P = 1.16×10-8), allergic asthma (OR = 1.44; 95% CI: 1.20-1.73; P = 8.63×10-5), and ILD (OR = 1.15; 95% CI: 1.04-1.26; P = 4.62×10-3). However, there was limited evidence to support an association between VAT and sarcoidosis. In multivariate MR analysis, VAT's associations with COPD, allergic asthma, and ILD persisted after adjusting for BMI. Conclusion This study provides evidence for a potential causal relationship between VAT and COPD, allergic asthma, and ILD; these relationships were independent of the effect of BMI.
Collapse
Affiliation(s)
- Jin-Xian Huang
- The Fourth Clinical Medicine College, Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Bing-Jie Xiao
- The Second Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Yu-Xin Yan
- The Fourth Clinical Medicine College, Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Wei Xie
- The Fourth Clinical Medicine College, Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Le-Yi Feng
- The Fourth Clinical Medicine College, Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Xue-Mei Liu
- Department of Endocrinology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, People’s Republic of China
| |
Collapse
|
2
|
Navarro-Ledesma S. Frozen Shoulder as a Metabolic and Immune Disorder: Potential Roles of Leptin Resistance, JAK-STAT Dysregulation, and Fibrosis. J Clin Med 2025; 14:1780. [PMID: 40095902 PMCID: PMC11901274 DOI: 10.3390/jcm14051780] [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: 01/21/2025] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
Frozen shoulder (FS) is a complex and multifactorial condition characterized by persistent inflammation, fibrosis, and metabolic dysregulation. Despite extensive research, the underlying drivers of FS remain poorly understood. Recent findings indicate the coexistence of pro-inflammatory and fibrosis-resolving macrophages within affected tissues, suggesting a dysregulated immune response influenced by metabolic and neuroendocrine factors. This review proposes that leptin resistance, a hallmark of metabolic syndrome and chronic inflammation, may play a central role in FS pathogenesis by impairing macrophage polarization, perpetuating inflammation, and disrupting fibrosis resolution. The JAK-STAT signaling pathway, critically modulated by leptin resistance, may further contribute to immune dysregulation by sustaining inflammatory macrophage activation and interfering with tissue remodeling. Additionally, FS shares pathogenic features with fibrotic diseases driven by TGF-β signaling, mitochondrial dysfunction, and circadian disruption, further linking systemic metabolic dysfunction to localized fibrotic pathology. Beyond immune and metabolic regulation, alterations in gut microbiota, bacterial translocation, and chronic psychosocial stress may further exacerbate systemic inflammation and neuroendocrine imbalances, intensifying JAK-STAT dysregulation and leptin resistance. By examining the intricate interplay between metabolism, immune function, and fibrotic remodeling, this review highlights targeting leptin sensitivity, JAK-STAT modulation, and mitochondrial restoration as novel therapeutic strategies for FS treatment. Future research should explore these interconnections to develop integrative interventions that address both the metabolic and immune dysregulation underlying FS, ultimately improving clinical outcomes.
Collapse
Affiliation(s)
- Santiago Navarro-Ledesma
- Department of Physiotherapy, Faculty of Health Sciences, Campus of Melilla, University of Granada, Querol Street 5, 52004 Melilla, Spain
| |
Collapse
|
3
|
Muñoz-Cofré R, Rojas-Mancilla E, Lizana PA, Escobar-Cabello M, García-Herrera C, Conei D, Valenzuela-Aedo F, Soto-Rodríguez FJ, del Sol M. Relationship Between Plasma Leptin Levels and Airflow Limitation in the Small and Medium Airways in Young Adults. J Clin Med 2025; 14:1624. [PMID: 40095561 PMCID: PMC11899985 DOI: 10.3390/jcm14051624] [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: 01/30/2025] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Background/Objectives: To determine the relationship between plasma leptin levels and airflow limitation (AFL) in the small and medium airways of young adults according to their body composition (BC). Methods: To determine AFL, the following measurements were taken: forced expiratory volume in the first second (FEV1), forced expiratory flow between 25-75% (FEF25-75%), airway resistance (Raw), and specific airway resistance (sRaw). The measured BC variables were body mass index (BMI), body fat percentage (%BF), trunk fat percentage (TF%), and fat-free mass (FFM). Plasma leptin was measured using the ELISA technique. Results: In total, 83 participants (40 male and 43 female) aged 21.55 ± 2.08 years were evaluated. Leptin was significantly higher in women than men (p < 0.01). When relating leptin to lung function variables, FEV1 and FEF25-75% showed a significant inverse relationship (p < 0.01), and Raw and sRaw showed a direct and significant relationship (p < 0.01). Female participants with a BF% and leptin higher than their p75 were observed to have a higher risk of increased sRaw (OR = 1.0; OR = 1.15, respectively), regardless of age, and participants with an FFM% higher than their p75 had a lower risk of increased sRaw (OR = 0.71), regardless of gender and age. Conclusions: There is an inverse relationship between FEV1, FEF25-75%, and a direct relationship between Raw and sRaw with leptin. For female participants with a higher BF%, higher FFM%, and leptin, the risk of developing Raw and sRaw was found to be modified.
Collapse
Affiliation(s)
- Rodrigo Muñoz-Cofré
- Ciencias Morfológicas, Universidad de La Frontera, Temuco 4811230, Chile; (F.V.-A.); (M.d.S.)
- Escuela de Kinesiología, Facultad de Salud, Universidad Santo Tomás, Manuel Rodríguez 060, Temuco 4801076, Chile
| | - Edgardo Rojas-Mancilla
- Unidad de Diagnóstico Laboratorio Clínico, Instituto Oncológico, Fundación Arturo López Pérez, Santiago 7500691, Chile;
- Escuela de Terapia Ocupacional, Facultad de Salud y Ciencias Sociales, Universidad de Las Américas, Santiago 7500975, Chile
| | - Pablo A. Lizana
- Laboratory of Epidemiology and Morphological Sciences, Instituto de Biología, Pontificia Universidad Católica de Valparaíso, Valparaíso 2373223, Chile;
| | - Máximo Escobar-Cabello
- Laboratorio de Función Disfunción Ventilatoria, Departamento de Kinesiología, Universidad Católica del Maule, Talca 3530000, Chile;
| | - Claudio García-Herrera
- Departamento de Ingeniería Mecánica, Universidad de Santiago de Chile, Santiago 9170022, Chile;
| | - Daniel Conei
- Departamento de Procesos Terapéuticos, Facultad de Ciencias de la Salud, Universidad Católica de Temuco, Temuco 4780000, Chile;
| | - Fernando Valenzuela-Aedo
- Ciencias Morfológicas, Universidad de La Frontera, Temuco 4811230, Chile; (F.V.-A.); (M.d.S.)
- Departamento de Ciencias de la Rehabilitación, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile;
| | - Francisco Javier Soto-Rodríguez
- Departamento de Ciencias de la Rehabilitación, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile;
- Programa de Doctorado en Medicina Clínica y Salud Pública, Universidad de Granada, 18071 Granada, Spain
| | - Mariano del Sol
- Ciencias Morfológicas, Universidad de La Frontera, Temuco 4811230, Chile; (F.V.-A.); (M.d.S.)
- Centro de Excelencia en Estudios Morfológicos y Quirúrgicos (CEMyQ), Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
| |
Collapse
|
4
|
Kaya H, Boyaci H, Argun Baris S, Basyigit I, Ozsoy OD, Maral Kir H. The predictive effects of adiponectin and irisin hormones on diagnosis and clinical involvement of Sarcoidosis. BMC Pulm Med 2024; 24:623. [PMID: 39695541 DOI: 10.1186/s12890-024-03412-z] [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: 08/27/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Sarcoidosis is a chronic disease of unknown etiology characterised by systemic non-caseating granulomas that can affect any organ in the body, especially the lungs and in which genetic and environmental factors are thought to play a role in its pathophysiology. Adipokines and myokines secreted from adipose and muscle tissue play a role in the pathogenesis or protection against many inflammatory and autoimmune diseases in which inflammation and immunity form the basis. In our study, we aimed to investigate the role of the irisin and adiponectin in sarcoidosis. METHODS The study included 90 sarcoidosis patients and 86 healthy subjects. Adiponectin and irisin levels were analysed in addition to standard tests for diagnosis and follow-up of patients with sarcoidosis. The sensitivity and specificity of serum irisin levels for the diagnosis of sarcoidosis were evaluate dusing ROC analysis. RESULTS Irisin levels were significantly lower in the patient group than in the control group (3.28-5.25, p < 0.001). There was no association between irisin levels and extrapulmonary involvement. The cut-off irisin value for the diagnosis of sarcoidosis was ≤ 4.2662 with 95% confidence interval, and the sensitivity and specificity were calculated as 84% and 55.8%, respectively. CONCLUSIONS To our knowledge, this is the first study to investigate irisin in sarcoidosis patients. Based on the available evidence, anti-inflammatory, anti-oxidant and anti-apoptocic effects of irisin may play a role in the pathophysiology of sarcoidosis. Although no significant difference was found in our study, we believe that a comprehensive evaluation of adiponectin in sarcoidosis is important.
Collapse
Affiliation(s)
- Huseyin Kaya
- Chest Diseases, Kocaeli City Hospital, Kocaeli, Turkey.
| | - Hasim Boyaci
- Chest Diseases, University of Kocaeli, Kocaeli, Turkey
| | | | | | - Ozgur Doga Ozsoy
- Department of Biochemistry, University of Kocaeli, Kocaeli, Turkey
| | - Hala Maral Kir
- Department of Biochemistry, University of Kocaeli, Kocaeli, Turkey
| |
Collapse
|
5
|
Luo X, Xiang F. Acute exacerbation of idiopathic pulmonary fibrosis a narrative review primary focus on treatments. J Thorac Dis 2024; 16:4727-4741. [PMID: 39144320 PMCID: PMC11320219 DOI: 10.21037/jtd-23-1565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 05/17/2024] [Indexed: 08/16/2024]
Abstract
Background and Objective Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrotic interstitial pneumonia, which is the commonest type of idiopathic interstitial pneumonia in the clinic. For most patients, the course of the disease is slow and prolonged, but a percentage of them develop an acute respiratory worsening during the disease, known as an acute exacerbation of IPF (AE-IPF). The updated guidelines define AE-IPF as an acute worsening of dyspnea in an IPF patient within 1 month and exclude other conditions such as left heart failure and pulmonary embolism. However, the prevention and treatment of AE-IPF are still unclear. Based on the high mortality rate caused by AE, in this article, we will focus on the latest research advances in AE-IPF treatment strategies and provide a comprehensive review of its pathogenesis, risk factors, clinical features, and diagnosis. Methods This study searched for relevant literature published from 2018 to 2023 in the PubMed database. The search terms used were as follows: "Acute exacerbation", "Idiopathic pulmonary fibrosis", "Biomarker", "Pathogenesis", "Treatment", "HRCT", "Antifibrotic", "Infection", "Immunosuppressant", "Autoantibody", "Oxygen therapy", "Hemoperfusion", "Inflammation". Key Content and Findings The review found that corticosteroids are still the primary treatment strategy at present, although there is some controversy regarding the dosing and tapering of corticosteroids. However, corticosteroids combined with intravenous cyclophosphamide have been shown to be detrimental to the prognosis of patients with AE-IPF. Given its deadly high mortality rate, early intervention is crucial. Pirfenidone and nintedanib have been proven to reduce incidence of AE. Meanwhile, in the future, the lung microbiome may also be a break-through. Conclusions This study reviewed the pathogenesis and risk factors of AE-IPF and updated the current and potential treatment strategies regarding AE-IPF. The pathogenesis of AE-IPF is not exact, multiple mechanisms may be involved simultaneously. Corticosteroids remain the mainstream treatment modality in the medical treatment of AE-IFP. Many other treatment modalities have been proposed in succession, but no clear conclusions can be drawn about the effectiveness and safety of these interventions.
Collapse
Affiliation(s)
- Xiaohui Luo
- Department of Pulmonary and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Xiang
- Department of Pulmonary and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Keller MB, Sun J, Alnababteh M, Ponor L, D. Shah P, Mathew J, Kong H, Charya A, Luikart H, Aryal S, Nathan SD, Orens JB, Khush KK, Kyoo Jang M, Agbor-Enoh S. Baseline Lung Allograft Dysfunction After Bilateral Lung Transplantation Is Associated With an Increased Risk of Death: Results From a Multicenter Cohort Study. Transplant Direct 2024; 10:e1669. [PMID: 38953039 PMCID: PMC11216668 DOI: 10.1097/txd.0000000000001669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 07/03/2024] Open
Abstract
Background A prior single-center, retrospective cohort study identified baseline lung allograft dysfunction (BLAD) as a risk factor for death in bilateral lung transplant recipients. In this multicenter prospective cohort study, we test the association of BLAD with death in bilateral lung transplant recipients, identify clinical risk factors for BLAD, and assess its association with allograft injury on the molecular level. Methods This multicenter, prospective cohort study included 173 bilateral lung transplant recipients that underwent serial pulmonary function testing and plasma collection for donor-derived cell-free DNA at prespecified time points. BLAD was defined as failure to achieve ≥80% predicted for both forced expiratory volume in 1 s and forced vital capacity after lung transplant, on 2 consecutive measurements at least 3 mo apart. Results BLAD was associated with increased risk of death (hazard ratio, 1.97; 95% confidence interval [CI], 1.05-3.69; P = 0.03) but not chronic lung allograft dysfunction alone (hazard ratio, 1.60; 95% CI, 0.87-2.95; P = 0.13). Recipient obesity (odds ratio, 1.69; 95% CI, 1.15-2.80; P = 0.04) and donor age (odds ratio, 1.03; 95% CI, 1.02-1.05; P = 0.004) increased the risk of developing BLAD. Patients with BLAD did not demonstrate higher log10(donor-derived cell-free DNA) levels compared with no BLAD (slope [SE]: -0.0095 [0.0007] versus -0.0109 [0.0007]; P = 0.15). Conclusions BLAD is associated with an increased risk of death following lung transplantation, representing an important posttransplant outcome with valuable prognostic significance; however, early allograft specific injury on the molecular level does not increase the risk of BLAD, supporting further mechanistic insight into disease pathophysiology.
Collapse
Affiliation(s)
- Michael B. Keller
- Laborarory of Applied Precision Omics (APO), National Institutes of Health, Bethesda, MD
- Laboratory of Transplantation Genomics, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Junfeng Sun
- Advanced Lung Disease Program and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, VA
| | - Muhtadi Alnababteh
- Laborarory of Applied Precision Omics (APO), National Institutes of Health, Bethesda, MD
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Lucia Ponor
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Division of Hospital Medicine, Johns Hopkins Bayview Medical Center, Baltimore, MD
| | - Pali D. Shah
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD
| | - Joby Mathew
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD
| | - Hyesik Kong
- Laborarory of Applied Precision Omics (APO), National Institutes of Health, Bethesda, MD
- Laboratory of Transplantation Genomics, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
| | - Ananth Charya
- Division of Pulmonary and Critical Care Medicine, University of Maryland Medical Center, Baltimore, MD
| | - Helen Luikart
- Genome Transplant Genomics (GTD), Stanford University School of Medicine, Palo Alto, CA
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, CA
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA
| | - Shambhu Aryal
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Advanced Lung Disease Program and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, VA
| | - Steven D. Nathan
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Advanced Lung Disease Program and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, VA
| | - Jonathan B. Orens
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD
| | - Kiran K. Khush
- Genome Transplant Genomics (GTD), Stanford University School of Medicine, Palo Alto, CA
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, CA
| | - Moon Kyoo Jang
- Laborarory of Applied Precision Omics (APO), National Institutes of Health, Bethesda, MD
- Laboratory of Transplantation Genomics, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
| | - Sean Agbor-Enoh
- Laborarory of Applied Precision Omics (APO), National Institutes of Health, Bethesda, MD
- Laboratory of Transplantation Genomics, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, MD
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, MD
| |
Collapse
|
7
|
Ye X, Zhang M, Gu H, Liu M, Zhao Y, Shi Y, Wu S, Jiang C, Ye X, Zhu H, Li Q, Huang X, Cao M. Animal models of acute exacerbation of pulmonary fibrosis. Respir Res 2023; 24:296. [PMID: 38007420 PMCID: PMC10675932 DOI: 10.1186/s12931-023-02595-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/07/2023] [Indexed: 11/27/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive scarring interstitial lung disease with an unknown cause. Some patients may experience acute exacerbations (AE), which result in severe lung damage visible on imaging or through examination of tissue samples, often leading to high mortality rates. However, the etiology and pathogenesis of AE-IPF remain unclear. AE-IPF patients exhibit diffuse lung damage, apoptosis of type II alveolar epithelial cells, and an excessive inflammatory response. Establishing a reliable animal model of AE is critical for investigating the pathogenesis. Recent studies have reported a variety of animal models for AE-IPF, each with its own advantages and disadvantages. These models are usually established in mice with bleomycin-induced pulmonary fibrosis, using viruses, bacteria, small peptides, or specific drugs. In this review, we present an overview of different AE models, hoping to provide a useful resource for exploring the mechanisms and targeted therapies for AE-IPF.
Collapse
Affiliation(s)
- Xu Ye
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Mingrui Zhang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huimin Gu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Mengying Liu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Yichao Zhao
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanchen Shi
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shufei Wu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Cheng Jiang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoling Ye
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Huihui Zhu
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qi Li
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinmei Huang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
- Nanjing Institute of Respiratory Diseases, Nanjing, China.
| | - Mengshu Cao
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Nanjing Institute of Respiratory Diseases, Nanjing, China.
| |
Collapse
|
8
|
Macklin M, Thompson C, Kawano-Dourado L, Bauer Ventura I, Weschenfelder C, Trostchansky A, Marcadenti A, Tighe RM. Linking Adiposity to Interstitial Lung Disease: The Role of the Dysfunctional Adipocyte and Inflammation. Cells 2023; 12:2206. [PMID: 37759429 PMCID: PMC10526202 DOI: 10.3390/cells12182206] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/19/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Adipose tissue has functions beyond its principal functions in energy storage, including endocrine and immune functions. When faced with a surplus of energy, the functions of adipose tissue expand by mechanisms that can be both adaptive and detrimental. These detrimental adipose tissue functions can alter normal hormonal signaling and promote local and systemic inflammation with wide-ranging consequences. Although the mechanisms by which adipose tissue triggers metabolic dysfunction and local inflammation have been well described, little is known about the relationship between adiposity and the pathogenesis of chronic lung conditions, such as interstitial lung disease (ILD). In this review, we detail the conditions and mechanisms by which adipose tissue becomes dysfunctional and relate this dysfunction to inflammatory changes observed in various forms of ILD. Finally, we review the existing basic and clinical science literature linking adiposity to ILD, highlighting the need for additional research on the mechanisms of adipocyte-mediated inflammation in ILD and its clinical implications.
Collapse
Affiliation(s)
- Michael Macklin
- Section of Rheumatology, The University of Chicago, Chicago, IL 60637, USA;
| | - Chelsea Thompson
- Section of Rheumatology, The University of Chicago, Chicago, IL 60637, USA;
| | - Leticia Kawano-Dourado
- Hcor Research Institute (IP-Hcor), Hcor, São Paulo 04004-050, Brazil; (L.K.-D.); (A.M.)
- Pulmonary Division, Heart Institute (InCor), University of Sao Paulo Medical School, São Paulo 05403-903, Brazil
| | | | - Camila Weschenfelder
- Graduate Program in Health Sciences (Cardiology), Cardiology Institute, University Foundation of Cardiology (IC/FUC), Porto Alegre 90050-170, Brazil;
| | - Andrés Trostchansky
- Department of Biochemistry and Biomedical Research Center, School of Medicine, University of the Republic, Montevideo 11800, Uruguay;
| | - Aline Marcadenti
- Hcor Research Institute (IP-Hcor), Hcor, São Paulo 04004-050, Brazil; (L.K.-D.); (A.M.)
- Graduate Program in Health Sciences (Cardiology), Cardiology Institute, University Foundation of Cardiology (IC/FUC), Porto Alegre 90050-170, Brazil;
- Graduate Program in Epidemiology, School of Public Health, University of São Paulo (FSP-USP), São Paulo 01246-904, Brazil
| | - Robert M. Tighe
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Durham, NC 27710, USA;
| |
Collapse
|
9
|
Cai Z, Zhong J, Jiang Y, Zhang J. Bariatric surgery and COVID-19 outcomes: a systematic review and meta-analysis. Surg Obes Relat Dis 2023; 19:1058-1066. [PMID: 37149418 PMCID: PMC10010835 DOI: 10.1016/j.soard.2023.02.023] [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: 11/20/2022] [Revised: 01/28/2023] [Accepted: 02/24/2023] [Indexed: 03/14/2023]
Abstract
BACKGROUND Obesity and its associated complications have a negative impact on human health. Metabolic and bariatric surgery (MBS) ameliorates a series of clinical manifestations associated with obesity. However, the overall efficacy of MBS on COVID-19 outcomes remains unclear. OBJECTIVES The objective of this article is to analyze the relationship between MBS and COVID-19 outcomes. SETTING A meta-analysis. METHODS The PubMed, Embase, Web of Science, and Cochrane Library databases were searched to retrieve the related articles from inception to December 2022. All original articles reporting MBS-confirmed SARS-CoV-2 infection were included. Outcomes including hospital admission, mortality, intensive care unit (ICU) admission, mechanical ventilation utilization, hemodialysis during admission, and hospital stay were selected. Meta-analysis with fixed or random-effect models was used and reported in terms of odds ratios (ORs) or weighted mean differences (WMDs) along with their 95% confidence intervals (CIs). Heterogeneity was assessed with the I2 test. Study quality was assessed using the Newcastle-Ottawa Scale. RESULTS A total of 10 clinical trials involving the investigation of 150,848 patients undergoing MBS interventions were included. Patients who underwent MBS had a lower risk of hospital admission (OR: .47, 95% CI: .34-.66, I2 = 0%), mortality (OR: .43, 95% CI: .28-.65, I2 = 63.6%), ICU admission (OR: .41, 95% CI: .21-.77, I2 = 0%), and mechanical ventilation (OR: .51, 95% CI: .35-.75, I2 = 56.2%) than those who did not undergo surgery, but MBS did not affect hemodialysis risk or COVID-19 infection rate. In addition, the length of hospital stay for patients with COVID-19 after MBS was significantly reduced (WMD: -1.81, 95% CI: -3.11-.52, I2 = 82.7%). CONCLUSIONS Our findings indicate that MBS is shown to improve COVID-19 outcomes, including hospital admission, mortality, ICU admission, mechanical ventilation, and hospital stay. Patients with obesity who have undergone MBS infected with COVID-19 will have better clinical outcomes than those without MBS.
Collapse
Affiliation(s)
- Zixin Cai
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jiaxin Zhong
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yingling Jiang
- Department of Metabolism and Endocrinology, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, Hunan, China
| | - Jingjing Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
10
|
Nigro E, D’Agnano V, Quarcio G, Mariniello DF, Bianco A, Daniele A, Perrotta F. Exploring the Network between Adipocytokines and Inflammatory Response in SARS-CoV-2 Infection: A Scoping Review. Nutrients 2023; 15:3806. [PMID: 37686837 PMCID: PMC10490077 DOI: 10.3390/nu15173806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/22/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Adipose tissue is actually regarded as an endocrine organ, rather than as an organ that merely stores energy. During the COVID-19 pandemic, obesity has undoubtedly emerged as one of the most important risk factors for disease severity and poor outcomes related to SARS-CoV-2 infection. The aberrant production of cytokine-like hormones, called adipokines, may contribute to alterations in metabolism, dysfunction in vascular endothelium and the creation of a state of general chronic inflammation. Moreover, chronic, low-grade inflammation linked to obesity predisposes the host to immunosuppression and excessive cytokine activation. In this respect, understanding the mechanisms that link obesity with the severity of SARS-CoV-2 infection could represent a real game changer in the development of new therapeutic strategies. Our review therefore examines the pathogenic mechanisms of SARS-CoV-2, the implications with visceral adipose tissue and the influences of the adipose tissue and its adipokines on the clinical behavior of COVID-19.
Collapse
Affiliation(s)
- Ersilia Nigro
- CEINGE-Biotecnologie Avanzate Scarl “Franco Salvatore”, Via G. Salvatore 486, 80145 Napoli, Italy; (E.N.); (A.D.)
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università della Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
| | - Vito D’Agnano
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80138 Naples, Italy; (V.D.); (G.Q.); (D.F.M.); (A.B.)
| | - Gianluca Quarcio
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80138 Naples, Italy; (V.D.); (G.Q.); (D.F.M.); (A.B.)
| | - Domenica Francesca Mariniello
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80138 Naples, Italy; (V.D.); (G.Q.); (D.F.M.); (A.B.)
| | - Andrea Bianco
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80138 Naples, Italy; (V.D.); (G.Q.); (D.F.M.); (A.B.)
| | - Aurora Daniele
- CEINGE-Biotecnologie Avanzate Scarl “Franco Salvatore”, Via G. Salvatore 486, 80145 Napoli, Italy; (E.N.); (A.D.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, 80055 Naples, Italy
| | - Fabio Perrotta
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80138 Naples, Italy; (V.D.); (G.Q.); (D.F.M.); (A.B.)
| |
Collapse
|
11
|
Huang D, Gong L, Wu Z, Shi Y, Liang Z. Genetic Association of Circulating Adipokines with Risk of Idiopathic Pulmonary Fibrosis: A Two-Sample Mendelian Randomization Study. Lung 2023; 201:355-362. [PMID: 37530803 DOI: 10.1007/s00408-023-00640-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023]
Abstract
PURPOSE The causal relationships between circulating adipokines and idiopathic pulmonary fibrosis (IPF) are yet to be established. We performed a two-sample Mendelian randomization (MR) study to investigate the causal roles of adipokines on IPF risk. METHODS We analyzed the summary data from genome-wide association studies (GWAS), including adiponectin, leptin, resistin and monocyte chemoattractant protein-1 (MCP-1) and IPF. The inverse-variance weighted (IVW) method was considered as the major method and the MR-Egger, weighted median, simple mode and weighted mode were utilized as complementary methods. We also performed the sensitivity analyses, including heterogeneity test, horizontal pleiotropy test and leave-one-out analysis. RESULTS The selected number of single nucleotide polymorphisms (SNPs) was 13 for adiponectin, 6 for leptin,12 for resistin, and 6 for MCP-1, respectively. The results showed a causal effect of the circulating adiponectin levels on the risk of IPF (OR 0.645, 95% CI 0.457-0.911, P = 0.013). However, we did not observe significant associations of genetic changes in serum leptin (OR 1.018, 95% CI 0.442-2.346, P = 0.967), resistin (OR 1.002, 95% CI 0.712-1.408, P = 0.993), and MCP-1 (OR 1.358, 95% CI 0.891-2.068, P = 0.155) with risk of developing IPF. There was no evidence of heterogeneity or horizontal pleiotropy. The sensitivity analyses confirmed that our results were stable and reliable. CONCLUSIONS The increase in serum adiponectin was associated causally with a decreased risk of developing IPF. There is no evidence to support a causal association between leptin, resistin or MCP-1 with risk of IPF. Further studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Dong Huang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Linjing Gong
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Zhenru Wu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Yujun Shi
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| | - Zongan Liang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
12
|
Yao S, Zeng L, Wang F, Chen K. Obesity Paradox in Lung Diseases: What Explains It? Obes Facts 2023; 16:411-426. [PMID: 37463570 PMCID: PMC10601679 DOI: 10.1159/000531792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/28/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Obesity is a globally increasing health problem that impacts multiple organ systems and a potentially modifiable risk factor for many diseases. Obesity has a significant impact on lung function and is strongly linked to the pathophysiology that contributes to lung diseases. On the other hand, reports have emerged that obesity is associated with a better prognosis than for normal weight individuals in some lung diseases, including pneumonia, acute lung injury/acute respiratory distress syndrome, chronic obstructive pulmonary disease, and lung cancer. The lesser mortality and better prognosis in patients with obesity is known as obesity paradox. While obesity paradox is both recognized and disputed in epidemiological studies, recent research has suggested possible mechanisms. SUMMARY In this review, we attempted to explain and summarize these factors and mechanisms, including immune response, pulmonary fibrosis, lung function, microbiota, fat and muscle reserves, which are significantly altered by obesity and may contribute to the obesity paradox in lung diseases. We also discuss contrary literature that attributes the "obesity paradox" to confounding. KEY MESSAGES The review will illustrate the possible role of obesity in the prognosis or course of lung diseases, leading to a better understanding of the obesity paradox and provide hints for further basic and clinical research in lung diseases.
Collapse
Affiliation(s)
- Surui Yao
- School of Public Health, Chengdu Medical College, Chengdu, PR China
| | - Lei Zeng
- School of Public Health, Chengdu Medical College, Chengdu, PR China
| | - Fengyuan Wang
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, PR China
| | - Kejie Chen
- School of Public Health, Chengdu Medical College, Chengdu, PR China
| |
Collapse
|
13
|
Avtaar Singh SS, Das De S, Al-Adhami A, Singh R, Hopkins PMA, Curry PA. Primary graft dysfunction following lung transplantation: From pathogenesis to future frontiers. World J Transplant 2023; 13:58-85. [PMID: 36968136 PMCID: PMC10037231 DOI: 10.5500/wjt.v13.i3.58] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/11/2022] [Accepted: 02/17/2023] [Indexed: 03/16/2023] Open
Abstract
Lung transplantation is the treatment of choice for patients with end-stage lung disease. Currently, just under 5000 lung transplants are performed worldwide annually. However, a major scourge leading to 90-d and 1-year mortality remains primary graft dysfunction. It is a spectrum of lung injury ranging from mild to severe depending on the level of hypoxaemia and lung injury post-transplant. This review aims to provide an in-depth analysis of the epidemiology, patho physiology, risk factors, outcomes, and future frontiers involved in mitigating primary graft dysfunction. The current diagnostic criteria are examined alongside changes from the previous definition. We also highlight the issues surrounding chronic lung allograft dysfunction and identify the novel therapies available for ex-vivo lung perfusion. Although primary graft dysfunction remains a significant contributor to 90-d and 1-year mortality, ongoing research and development abreast with current technological advancements have shed some light on the issue in pursuit of future diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Sanjeet Singh Avtaar Singh
- Department of Cardiothoracic Surgery, Royal Infirmary of Edinburgh, Edinburgh EH16 4SA, United Kingdom
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Sudeep Das De
- Heart and Lung Transplant Unit, Wythenshawe Hospital, Manchester M23 9NJ, United Kingdom
| | - Ahmed Al-Adhami
- Department of Cardiothoracic Surgery, Royal Infirmary of Edinburgh, Edinburgh EH16 4SA, United Kingdom
- Department of Heart and Lung Transplant, Royal Papworth Hospital, Cambridge CB2 0AY, United Kingdom
| | - Ramesh Singh
- Mechanical Circulatory Support, Inova Health System, Falls Church, VA 22042, United States
| | - Peter MA Hopkins
- Queensland Lung Transplant Service, Prince Charles Hospital, Brisbane, QLD 4032, Australia
| | - Philip Alan Curry
- Department of Cardiothoracic Surgery, Golden Jubilee National Hospital, Glasgow G81 4DY, United Kingdom
| |
Collapse
|
14
|
El Husseini K, Poté N, Jaillet M, Mordant P, Mal H, Frija-Masson J, Borie R, Cazes A, Crestani B, Mailleux A. [Adipocytes, adipokines and metabolic alterations in pulmonary fibrosis]. Rev Mal Respir 2023; 40:225-229. [PMID: 36740493 DOI: 10.1016/j.rmr.2023.01.016] [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: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 02/07/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal respiratory disease characterized by severe remodeling of the lung parenchyma, with an accumulation of activated myofibroblasts and extracellular matrix, along with aberrant cellular differentiation. Within the subpleural fibrous zones, ectopic adipocyte deposits often appear. In addition, alterations in lipid homeostasis have been associated with IPF pathophysiology. In this mini-review, we will discuss the potential involvement of the adipocyte secretome and its paracrine or endocrine-based contribution to the pathophysiology of IPF, via protein or lipid mediators in particular.
Collapse
Affiliation(s)
- K El Husseini
- Service de pneumologie A, Hôpital Bichat, AP-HP ; Inserm Unit 1152, Université de Paris, Paris, France; Inserm Unité 1152 - PHERE, Université de Paris, Paris, France.
| | - N Poté
- Service d'anatomopathologie, Hôpital Bichat, AP-HP ; Inserm Unité 1152 - PHERE, Université de Paris, Paris, France
| | - M Jaillet
- Inserm Unité 1152 - PHERE, Université de Paris, Paris, France
| | - P Mordant
- Service de chirurgie vasculaire et thoracique, Hôpital Bichat, AP-HP, Paris, France
| | - H Mal
- Service de pneumologie B, Hôpital Bichat, AP-HP ; Inserm Unité 1152 - PHERE, Université de Paris, Paris, France
| | - J Frija-Masson
- Service de physiologie-explorations fonctionnelles respiratoires, Hôpital Bichat, AP-HP, Paris, France
| | - R Borie
- Service de pneumologie A, Hôpital Bichat, AP-HP ; Inserm Unit 1152, Université de Paris, Paris, France
| | - A Cazes
- Service d'anatomopathologie, Hôpital Bichat, AP-HP ; Inserm Unité 1152 - PHERE, Université de Paris, Paris, France
| | - B Crestani
- Service de pneumologie A, Hôpital Bichat, AP-HP ; Inserm Unit 1152, Université de Paris, Paris, France; Inserm Unité 1152 - PHERE, Université de Paris, Paris, France
| | - A Mailleux
- Inserm Unité 1152 - PHERE, Université de Paris, Paris, France
| |
Collapse
|
15
|
Wang Y, Hu C. Leptin and Asthma: What Are the Interactive Correlations? Biomolecules 2022; 12:biom12121780. [PMID: 36551211 PMCID: PMC9775505 DOI: 10.3390/biom12121780] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Leptin is an adipokine directly correlated with the proinflammatory obese-associated phenotype. Leptin has been demonstrated to inhibit adipogenesis, promote fat demarcation, promote a chronic inflammatory state, increase insulin sensitivity, and promote angiogenesis. Leptin, a regulator of the immune response, is implicated in the pathology of asthma. Studies involved in the key cell reaction and animal models of asthma have provided vital insights into the proinflammatory role of leptin in asthma. Many studies described the immune cell and related cellular pathways activated by leptin, which are beneficial in asthma development and increasing exacerbations. Subsequent studies relating to animal models support the role of leptin in increasing inflammatory cell infiltration, airway hyperresponsiveness, and inflammatory responses. However, the conclusive effects of leptin in asthma are not well elaborated. In the present study, we explored the general functions and the clinical cohort study supporting the association between leptin and asthma. The main objective of our review is to address the knowns and unknowns of leptin on asthma. In this perspective, the arguments about the different faces of leptin in asthma are provided to picture the potential directions, thus yielding a better understanding of asthma development.
Collapse
Affiliation(s)
- Yang Wang
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chengping Hu
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Xiangya Hospital, Central South University, Changsha 410008, China
- Correspondence:
| |
Collapse
|
16
|
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has brought severe challenges to global public health. Many studies have shown that obesity plays a vital role in the occurrence and development of COVID-19. Obesity exacerbates COVID-19, leading to increased intensive care unit hospitalization rate, high demand for invasive mechanical ventilation, and high mortality. The mechanisms of interaction between obesity and COVID-19 involve inflammation, immune response, changes in pulmonary dynamics, disruptions of receptor ligands, and dysfunction of endothelial cells. Therefore, for obese patients with COVID-19, the degree of obesity and related comorbidities should be evaluated. Treatment methods such as administration of anticoagulants and anti-inflammatory drugs like glucocorticoids and airway management should be actively initiated. We should also pay attention to long-term prognosis and vaccine immunity and actively address the physical and psychological problems caused by longterm staying-at-home during the pandemic. The present study summarized the research to investigate the role of obesity in the incidence and progression of COVID-19 and the psychosocial impact and treatment options for obese patients with COVID-19, to guide the understanding and management of the disease.
Collapse
Affiliation(s)
- Sijia Fei
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing100730, China
- Graduate School of Peking Union Medical College, Beijing100730, China
| | - Xinyuan Feng
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing100730, China
- Graduate School of Peking Union Medical College, Beijing100730, China
| | - Jingyi Luo
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing100730, China
| | - Lixin Guo
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing100730, China
| | - Qi Pan
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing100730, China
- Graduate School of Peking Union Medical College, Beijing100730, China
| |
Collapse
|
17
|
Ihrie MD, McQuade VL, Womble JT, Hegde A, McCravy MS, Lacuesta CVG, Tighe RM, Que LG, Walker JKL, Ingram JL. Exogenous leptin enhances markers of airway fibrosis in a mouse model of chronic allergic airways disease. Respir Res 2022; 23:131. [PMID: 35610699 PMCID: PMC9131622 DOI: 10.1186/s12931-022-02048-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Asthma patients with comorbid obesity exhibit increased disease severity, in part, due to airway remodeling, which is also observed in mouse models of asthma and obesity. A mediator of remodeling that is increased in obesity is leptin. We hypothesized that in a mouse model of allergic airways disease, mice receiving exogenous leptin would display increased airway inflammation and fibrosis. METHODS Five-week-old male and female C57BL/6J mice were challenged with intranasal house dust mite (HDM) allergen or saline 5 days per week for 6 weeks (n = 6-9 per sex, per group). Following each HDM exposure, mice received subcutaneous recombinant human leptin or saline. At 48 h after the final HDM challenge, lung mechanics were evaluated and the mice were sacrificed. Bronchoalveolar lavage was performed and differential cell counts were determined. Lung tissue was stained with Masson's trichrome, periodic acid-Schiff, and hematoxylin and eosin stains. Mouse lung fibroblasts were cultured, and whole lung mRNA was isolated. RESULTS Leptin did not affect mouse body weight, but HDM+leptin increased baseline blood glucose. In mixed-sex groups, leptin increased mouse lung fibroblast invasiveness and increased lung Col1a1 mRNA expression. Total lung resistance and tissue damping were increased with HDM+leptin treatment, but not leptin or HDM alone. Female mice exhibited enhanced airway responsiveness to methacholine with HDM+leptin treatment, while leptin alone decreased total respiratory system resistance in male mice. CONCLUSIONS In HDM-induced allergic airways disease, administration of exogenous leptin to mice enhanced lung resistance and increased markers of fibrosis, with differing effects between males and females.
Collapse
Affiliation(s)
- Mark D Ihrie
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University, Durham, NC, USA
| | - Victoria L McQuade
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University, Durham, NC, USA
| | - Jack T Womble
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University, Durham, NC, USA
| | - Akhil Hegde
- School of Nursing, Duke University, Durham, NC, USA
| | - Matthew S McCravy
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University, Durham, NC, USA
| | | | - Robert M Tighe
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University, Durham, NC, USA
| | - Loretta G Que
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University, Durham, NC, USA
| | - Julia K L Walker
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University, Durham, NC, USA
- School of Nursing, Duke University, Durham, NC, USA
| | - Jennifer L Ingram
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University, Durham, NC, USA.
- , Durham, USA.
| |
Collapse
|
18
|
Shchepikhin EI, Shmelev EI, Zaytseva AS. Respiratory diseases and obesity: special phenotype or independent events: Review. TERAPEVT ARKH 2022; 94:442-447. [DOI: 10.26442/00403660.2022.03.201412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/18/2022] [Indexed: 11/22/2022]
Abstract
A combination of factors, including Western European eating habits, physical inactivity and genetic predisposition, lead to a dramatic increase in adipose tissue mass. A special place is occupied by abdominal obesity, in which there is an accumulation of adipose tissue in the mesentery of the small intestine and the omentum. Developing in conditions of visceral obesity, insulin resistance, dyslipidemia and systemic inflammation are one of the key components of the pathogenesis of type 2 diabetes mellitus, cardiovascular diseases, non-alcoholic fatty liver and pancreas disease, polycystic ovary disease, some forms of cancer (breast cancer, endometrial cancer, colonic and direct intestines). At the same time, the pathogenetic role of adipose tissue is not limited to its participation in the formation of the cardiometabolic continuum and oncogenesis. The most important role of metabolically active fat in the pathogenesis of many respiratory diseases is known, including bronchial asthma, obstructive sleep apnea and pulmonary hypertension. This paper presents an overview of current data on immunological, pathophysiological and clinical features of the phenotype of the combination of respiratory diseases with overweight and obesity.
Collapse
|
19
|
Mechanisms contributing to adverse outcomes of COVID-19 in obesity. Mol Cell Biochem 2022; 477:1155-1193. [PMID: 35084674 PMCID: PMC8793096 DOI: 10.1007/s11010-022-04356-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/07/2022] [Indexed: 01/08/2023]
Abstract
A growing amount of epidemiological data from multiple countries indicate an increased prevalence of obesity, more importantly central obesity, among hospitalized subjects with COVID-19. This suggests that obesity is a major factor contributing to adverse outcome of the disease. As it is a metabolic disorder with dysregulated immune and endocrine function, it is logical that dysfunctional metabolism contributes to the mechanisms behind obesity being a risk factor for adverse outcome in COVID-19. Emerging data suggest that in obese subjects, (a) the molecular mechanisms of viral entry and spread mediated through ACE2 receptor, a multifunctional host cell protein which links to cellular homeostasis mechanisms, are affected. This includes perturbation of the physiological renin-angiotensin system pathway causing pro-inflammatory and pro-thrombotic challenges (b) existent metabolic overload and ER stress-induced UPR pathway make obese subjects vulnerable to severe COVID-19, (c) host cell response is altered involving reprogramming of metabolism and epigenetic mechanisms involving microRNAs in line with changes in obesity, and (d) adiposopathy with altered endocrine, adipokine, and cytokine profile contributes to altered immune cell metabolism, systemic inflammation, and vascular endothelial dysfunction, exacerbating COVID-19 pathology. In this review, we have examined the available literature on the underlying mechanisms contributing to obesity being a risk for adverse outcome in COVID-19.
Collapse
|
20
|
von Knethen A, Heinicke U, Laux V, Parnham MJ, Steinbicker AU, Zacharowski K. Antioxidants as Therapeutic Agents in Acute Respiratory Distress Syndrome (ARDS) Treatment-From Mice to Men. Biomedicines 2022; 10:98. [PMID: 35052778 PMCID: PMC8773193 DOI: 10.3390/biomedicines10010098] [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: 12/09/2021] [Revised: 12/26/2021] [Accepted: 12/31/2021] [Indexed: 12/16/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a major cause of patient mortality in intensive care units (ICUs) worldwide. Considering that no causative treatment but only symptomatic care is available, it is obvious that there is a high unmet medical need for a new therapeutic concept. One reason for a missing etiologic therapy strategy is the multifactorial origin of ARDS, which leads to a large heterogeneity of patients. This review summarizes the various kinds of ARDS onset with a special focus on the role of reactive oxygen species (ROS), which are generally linked to ARDS development and progression. Taking a closer look at the data which already have been established in mouse models, this review finally proposes the translation of these results on successful antioxidant use in a personalized approach to the ICU patient as a potential adjuvant to standard ARDS treatment.
Collapse
Affiliation(s)
- Andreas von Knethen
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | - Ulrike Heinicke
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Volker Laux
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | - Michael J Parnham
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | - Andrea U Steinbicker
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Kai Zacharowski
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
21
|
Valera RJ, Botero-Fonnegra C, Cogollo VJ, Montorfano L, Sarmiento-Cobos M, Rivera CE, Hong L, Lo Menzo E, Szomstein S, Rosenthal RJ. Impact of bariatric surgery on the risk of hospitalization due to influenza virus infection. Surg Obes Relat Dis 2021; 17:1977-1983. [PMID: 34593336 DOI: 10.1016/j.soard.2021.08.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/06/2021] [Accepted: 08/17/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Obesity independently increases the risk of hospitalization due to viral respiratory infections, including influenza virus and, more recently, severe acute respiratory syndrome coronavirus 2. As an independent risk factor, obesity impairs the immune response to viral infections and decreases the effectiveness of immunizations. OBJECTIVES Using influenza as a proxy, we aimed to determine the impact of bariatric surgery (BaS) on the risk of hospitalization due to viral respiratory infections. SETTING Academic hospital, United States. METHODS National (Nationwide) Inpatient Sample data collected from 2010 to 2015 were examined. Patients were classified as treatment and control groups. Treatment subjects were defined as patients with a history of BaS and control subjects as patients with a body mass index ≥35 kg/m2 and without a history of BaS. Any hospitalization with influenza as a primary diagnosis was identified. Univariate analysis and multivariate regression models were performed to assess the differences between groups. RESULTS A total of 2,300,845 subjects were reviewed, of which 2,004,804 were control subjects and 296,041 were treated patients. Univariate analysis showed that the hospitalization rate in the treatment group was significantly lower than in the control group (.007% versus .019%, P < .0001), which was confirmed after adjusting for covariables (control versus treatment: odds ratio = 2.21, P = .0010). CONCLUSIONS BaS may decrease the risk of hospitalization due to influenza, but further prospective studies are needed to confirm these results. We also suggest that these results should be translated into the development of similar studies to determine the impact of BaS on the incidence and severity of the coronavirus disease 2019.
Collapse
Affiliation(s)
- Roberto J Valera
- Department of General Surgery and the Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, Florida
| | - Cristina Botero-Fonnegra
- Department of General Surgery and the Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, Florida
| | - Vicente J Cogollo
- Department of General Surgery, Kendall Regional Medical Center, Miami, Florida
| | - Lisandro Montorfano
- Department of General Surgery and the Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, Florida
| | - Mauricio Sarmiento-Cobos
- Department of General Surgery and the Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, Florida
| | - Carlos E Rivera
- Department of General Surgery and the Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, Florida
| | - Liang Hong
- Department of General Surgery and the Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, Florida
| | - Emanuele Lo Menzo
- Department of General Surgery and the Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, Florida
| | - Samuel Szomstein
- Department of General Surgery and the Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, Florida
| | - Raul J Rosenthal
- Department of General Surgery and the Bariatric and Metabolic Institute, Cleveland Clinic Florida, Weston, Florida.
| |
Collapse
|
22
|
Proteomics and metabonomics analyses of Covid-19 complications in patients with pulmonary fibrosis. Sci Rep 2021; 11:14601. [PMID: 34272434 PMCID: PMC8285535 DOI: 10.1038/s41598-021-94256-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 06/29/2021] [Indexed: 12/23/2022] Open
Abstract
Pulmonary fibrosis is a devastating disease, and the pathogenesis of this disease is not completely clear. Here, the medical records of 85 Covid-19 cases were collected, among which fibrosis and progression of fibrosis were analyzed in detail. Next, data independent acquisition (DIA) quantification proteomics and untargeted metabolomics were used to screen disease-related signaling pathways through clustering and enrichment analysis of the differential expression of proteins and metabolites. The main imaging features were lesions located in the bilateral lower lobes and involvement in five lobes. The closed association pathways were FcγR-mediated phagocytosis, PPAR signaling, TRP-inflammatory pathways, and the urea cycle. Our results provide evidence for the detection of serum biomarkers and targeted therapy in patients with Covid-19.
Collapse
|
23
|
Marchesi F, Valente M, Riccò M, Rottoli M, Baldini E, Mecheri F, Bonilauri S, Boschi S, Bernante P, Sciannamea A, Rolla J, Francescato A, Bollino R, Cartelli C, Lanaia A, Anzolin F, Del Rio P, Fabbi D, Petracca GL, Tartamella F, Dalmonte G. Effects of Bariatric Surgery on COVID-19: a Multicentric Study from a High Incidence Area. Obes Surg 2021; 31:2477-2488. [PMID: 33417099 PMCID: PMC7791147 DOI: 10.1007/s11695-020-05193-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/01/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The favorable effects of bariatric surgery (BS) on overall pulmonary function and obesity-related comorbidities could influence SARS-CoV-2 clinical expression. This has been investigated comparing COVID-19 incidence and clinical course between a cohort of patients submitted to BS and a cohort of candidates for BS during the spring outbreak in Italy. MATERIALS AND METHODS From April to August 2020, 594 patients from 6 major bariatric centers in Emilia-Romagna were administered an 87-item telephonic questionnaire. Demographics, COVID-19 incidence, suggestive symptoms, and clinical outcome parameters of operated patients and candidates to BS were compared. The incidence of symptomatic COVID-19 was assessed including the clinical definition of probable case, according to World Health Organization criteria. RESULTS Three hundred fifty-three operated patients (Op) and 169 candidates for BS (C) were finally included in the statistical analysis. While COVID-19 incidence confirmed by laboratory tests was similar in the two groups (5.7% vs 5.9%), lower incidence of most of COVID-19-related symptoms, such as anosmia (p: 0.046), dysgeusia (p: 0.049), fever with rapid onset (p: 0.046) were recorded among Op patients, resulting in a lower rate of probable cases (14.4% vs 23.7%; p: 0.009). Hospitalization was more frequent in C patients (2.4% vs 0.3%, p: 0.02). One death in each group was reported (0.3% vs 0.6%). Previous pneumonia and malignancies resulted to be associated with symptomatic COVID-19 at univariate and multivariate analysis. CONCLUSION Patients submitted to BS seem to develop less severe SARS-CoV-2 infection than subjects suffering from obesity.
Collapse
Affiliation(s)
- Federico Marchesi
- Unit of General Surgery, Parma University Hospital, Parma, Italy.
- Università degli Studi di Parma, Via Gramsci, 14-43126, Parma, Italy.
| | - Marina Valente
- Unit of General Surgery, Parma University Hospital, Parma, Italy
| | - Matteo Riccò
- Dipartimento di Sanità Pubblica/Public Health, AUSL-IRCCS Tecnologie Avanzate e Modelli Assistenziali in Oncologia di Reggio Emilia, Reggio Emilia, Italy
| | - Matteo Rottoli
- Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, Bologna, Italy
- Centre for the Study and Research of Treatment for Morbid Obesity, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Edoardo Baldini
- Department of Surgery, Ospedale "Guglielmo da Saliceto", Piacenza, Italy
| | - Fouzia Mecheri
- Division of General, Emergency Surgery and New Technologies, OCSAE (Ospedale Civile Sant'Agostino Estense), Baggiovara, Modena, Italy
| | - Stefano Bonilauri
- General and Emergency Surgery Unit, Arcispedale Santa Maria Nuova di Reggio Emilia, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Sergio Boschi
- Programma Dipartimentale Chirurgia Malassorbitiva AUSL di Bologna, Bologna, Italy
| | - Paolo Bernante
- Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, Bologna, Italy
- Centre for the Study and Research of Treatment for Morbid Obesity, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Andrea Sciannamea
- Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, Bologna, Italy
| | - Jessica Rolla
- Department of Medicine, Ospedale "Guglielmo da Saliceto", Piacenza, Italy
| | - Alice Francescato
- Division of General, Emergency Surgery and New Technologies, OCSAE (Ospedale Civile Sant'Agostino Estense), Baggiovara, Modena, Italy
| | - Ruggero Bollino
- General and Emergency Surgery Unit, Arcispedale Santa Maria Nuova di Reggio Emilia, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Concetto Cartelli
- General and Emergency Surgery Unit, Arcispedale Santa Maria Nuova di Reggio Emilia, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Andrea Lanaia
- General and Emergency Surgery Unit, Arcispedale Santa Maria Nuova di Reggio Emilia, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Francesca Anzolin
- Medical Department, Clinical Nutrition Unit, Maggiore-Bentivoglio Hospital, Ausl Bologna, Bologna, Italy
| | - Paolo Del Rio
- Unit of General Surgery, Parma University Hospital, Parma, Italy
| | - Diletta Fabbi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | | | - Giorgio Dalmonte
- Unit of General Surgery, Parma University Hospital, Parma, Italy
| |
Collapse
|
24
|
McNeill JN, Lau ES, Zern EK, Nayor M, Malhotra R, Liu EE, Bhat RR, Brooks LC, Farrell R, Sbarbaro JA, Schoenike MW, Medoff BD, Lewis GD, Ho JE. Association of obesity-related inflammatory pathways with lung function and exercise capacity. Respir Med 2021; 183:106434. [PMID: 33964816 DOI: 10.1016/j.rmed.2021.106434] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND Obesity has multifactorial effects on lung function and exercise capacity. The contributions of obesity-related inflammatory pathways to alterations in lung function remain unclear. RESEARCH QUESTION To examine the association of obesity-related inflammatory pathways with pulmonary function, exercise capacity, and pulmonary-specific contributors to exercise intolerance. METHOD We examined 695 patients who underwent cardiopulmonary exercise testing (CPET) with invasive hemodynamic monitoring at Massachusetts General Hospital between December 2006-June 2017. We investigated the association of adiponectin, leptin, resistin, IL-6, CRP, and insulin resistance (HOMA-IR) with pulmonary function and exercise parameters using multivariable linear regression. RESULTS Obesity-related inflammatory pathways were associated with worse lung function. Specifically, higher CRP, IL-6, and HOMA-IR were associated with lower percent predicted FEV1 and FVC with a preserved FEV1/FVC ratio suggesting a restrictive physiology pattern (P ≤ 0.001 for all). For example, a 1-SD higher natural-logged CRP level was associated with a nearly 5% lower percent predicted FEV1 and FVC (beta -4.8, s.e. 0.9 for FEV1; beta -4.9, s.e. 0.8 for FVC; P < 0.0001 for both). Obesity-related inflammatory pathways were associated with worse pulmonary vascular distensibility (adiponectin, IL-6, and CRP, P < 0.05 for all), as well as lower pulmonary artery compliance (IL-6 and CRP, P ≤ 0.01 for both). INTERPRETATION Our findings highlight the importance of obesity-related inflammatory pathways including inflammation and insulin resistance on pulmonary spirometry and pulmonary vascular function. Specifically, systemic inflammation as ascertained by CRP, IL-6 and insulin resistance are associated with restrictive pulmonary physiology independent of BMI. In addition, inflammatory markers were associated with lower exercise capacity and pulmonary vascular dysfunction.
Collapse
Affiliation(s)
- Jenna N McNeill
- From the Cardiovascular Research Center, Division of Massachusetts General Hospital, Boston, MA, USA; Pulmonary and Critical Care, Division of Massachusetts General Hospital, Boston, MA, USA
| | - Emily S Lau
- From the Cardiovascular Research Center, Division of Massachusetts General Hospital, Boston, MA, USA; Cardiology Division of Massachusetts General Hospital, Boston, MA, USA
| | - Emily K Zern
- From the Cardiovascular Research Center, Division of Massachusetts General Hospital, Boston, MA, USA; Cardiology Division of Massachusetts General Hospital, Boston, MA, USA
| | - Matthew Nayor
- Cardiology Division of Massachusetts General Hospital, Boston, MA, USA
| | - Rajeev Malhotra
- Cardiology Division of Massachusetts General Hospital, Boston, MA, USA
| | - Elizabeth E Liu
- From the Cardiovascular Research Center, Division of Massachusetts General Hospital, Boston, MA, USA
| | - Rohan R Bhat
- Cardiology Division of Massachusetts General Hospital, Boston, MA, USA
| | - Liana C Brooks
- Cardiology Division of Massachusetts General Hospital, Boston, MA, USA
| | - Robyn Farrell
- Cardiology Division of Massachusetts General Hospital, Boston, MA, USA
| | - John A Sbarbaro
- Cardiology Division of Massachusetts General Hospital, Boston, MA, USA
| | - Mark W Schoenike
- Cardiology Division of Massachusetts General Hospital, Boston, MA, USA
| | - Benjamin D Medoff
- Pulmonary and Critical Care, Division of Massachusetts General Hospital, Boston, MA, USA
| | - Gregory D Lewis
- Cardiology Division of Massachusetts General Hospital, Boston, MA, USA
| | - Jennifer E Ho
- From the Cardiovascular Research Center, Division of Massachusetts General Hospital, Boston, MA, USA; Cardiology Division of Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
25
|
Anderson MR, Kim JS, Allison M, Giles JT, Hoffman EA, Ding J, Barr RG, Podolanczuk A. Adiposity and Interstitial Lung Abnormalities in Community-Dwelling Adults: The MESA Cohort Study. Chest 2021; 160:582-594. [PMID: 33844978 DOI: 10.1016/j.chest.2021.03.058] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/13/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Obesity is associated with restrictive ventilatory defects and a faster rate of decline in FVC. This association is not exclusively mediated by mechanical factors and may reflect direct pulmonary injury by adipose-derived mediators. RESEARCH QUESTION Is adipose tissue involved in the pathogenesis of interstitial lung disease (ILD)? STUDY DESIGN AND METHODS We evaluated the association of CT measures of pericardial, abdominal visceral, and abdominal subcutaneous adipose tissue with high-attenuation areas (HAAs) and interstitial lung abnormalities (ILAs) in a large multicenter cohort study of community-dwelling adults, using multivariable-adjusted models. We secondarily evaluated the association of adipose depot size with FVC and biomarkers of obesity and inflammation. RESULTS In fully adjusted models, every doubling in pericardial adipose tissue volume was associated with a 63.4-unit increase in HAA (95% CI, 55.5-71.3), 20% increased odds of ILA (95% CI, -2% to 50%), and a 5.5% decrease in percent predicted FVC (95% CI, -6.8% to -4.3%). IL-6 levels accounted for 8% of the association between pericardial adipose tissue and HAA. Every doubling in visceral adipose tissue area was associated with a 41.5-unit increase in HAA (95% CI, 28.3-54.7), 30% increased odds of ILA (95% CI, -10% to 80%), and a 5.4% decrease in percent predicted FVC (95% CI, -6.6% to -4.3%). IL-6 and leptin accounted for 17% and 18%, respectively, of the association between visceral adipose tissue and HAA. INTERPRETATION Greater amounts of pericardial and abdominal visceral adipose tissue were associated with CT measures of early lung injury and lower FVC in a cohort of community-dwelling adults. Adipose tissue may represent a modifiable risk factor for ILD.
Collapse
Affiliation(s)
| | - John S Kim
- Department of Medicine, University of Virginia, Charlottesville, VA
| | - Matthew Allison
- Department of Preventive Medicine, University of California San Diego, San Diego, CA
| | - Jon T Giles
- Department of Medicine, Columbia University Medical Center, New York, NY
| | - Eric A Hoffman
- Department of Radiology, University of Iowa, Des Moines, IA
| | - Jingzhong Ding
- Department of Epidemiology and Prevention, Wake Forest University, Winston-Salem, NC; Department of Gerontology and Geriatric Science, Wake Forest University, Winston-Salem, NC
| | - R Graham Barr
- Department of Medicine, Columbia University Medical Center, New York, NY; Department of Epidemiology, Columbia University Medical Center, New York, NY
| | - Anna Podolanczuk
- Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
26
|
O’Rourke RW, Lumeng CN. Pathways to Severe COVID-19 for People with Obesity. Obesity (Silver Spring) 2021; 29:645-653. [PMID: 33270351 PMCID: PMC7753541 DOI: 10.1002/oby.23099] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 12/01/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022]
Abstract
Increased morbidity and mortality from coronavirus disease 2019 (COVID-19) in people with obesity have illuminated the intersection of obesity with impaired responses to infections. Although data on mechanisms by which COVID-19 impacts health are being rapidly generated, there is a critical need to better understand the pulmonary, vascular, metabolic, and immunologic aspects that drive the increased risk for complications from COVID-19 in people with obesity. This review provides a broad overview of the intersection between COVID-19 and the physiology of obesity in order to highlight potential mechanisms by which COVID-19 disease severity is increased by obesity and identify areas for future investigation toward developing tailored therapy for people with obesity who develop COVID-19.
Collapse
Affiliation(s)
- Robert W. O’Rourke
- Department of SurgeryUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
- Department of SurgeryAnn Arbor Veterans Affairs Healthcare SystemAnn ArborMichiganUSA
| | - Carey N. Lumeng
- Division of Pediatric PulmonologyDepartment of PediatricsUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| |
Collapse
|
27
|
Jutant EM, Tu L, Humbert M, Guignabert C, Huertas A. The Thousand Faces of Leptin in the Lung. Chest 2020; 159:239-248. [PMID: 32795478 DOI: 10.1016/j.chest.2020.07.075] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/21/2022] Open
Abstract
Leptin is a pleotropic hormone known to regulate a wide range of systemic functions, from satiety to inflammation. Increasing evidence has shown that leptin and its receptor (ObR) are not only expressed in adipose tissue but also in several organs, including the lungs. Leptin levels were first believed to be elevated only in the lungs of obese patients, and leptin was suspected to be responsible for obesity-related lung complications. Aside from obesity, leptin displays many faces in the respiratory system, independently of body weight, as this cytokine-like hormone plays important physiological roles, from the embryogenic state to maturation of the lungs and the control of ventilation. The leptin-signaling pathway is also involved in immune modulation and cell proliferation, and its dysregulation can lead to the onset of lung diseases. This review article addresses the thousand faces of leptin and its signaling in the lungs under physiological conditions and in disease.
Collapse
Affiliation(s)
- Etienne-Marie Jutant
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, "Pulmonary Hypertension: Pathophysiology and Novel Therapies," Hôpital Marie Lannelongue, Le Plessis-Robinson, France; Assistance Publique-Hôpitaux de Paris, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Ly Tu
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, "Pulmonary Hypertension: Pathophysiology and Novel Therapies," Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Marc Humbert
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, "Pulmonary Hypertension: Pathophysiology and Novel Therapies," Hôpital Marie Lannelongue, Le Plessis-Robinson, France; Assistance Publique-Hôpitaux de Paris, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, "Pulmonary Hypertension: Pathophysiology and Novel Therapies," Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Alice Huertas
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, "Pulmonary Hypertension: Pathophysiology and Novel Therapies," Hôpital Marie Lannelongue, Le Plessis-Robinson, France; Assistance Publique-Hôpitaux de Paris, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.
| |
Collapse
|
28
|
Electroacupuncture Pretreatment Alleviates LPS-Induced Acute Respiratory Distress Syndrome via Regulating the PPAR Gamma/NF-Kappa B Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:4594631. [PMID: 32774418 PMCID: PMC7396021 DOI: 10.1155/2020/4594631] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/27/2020] [Accepted: 06/30/2020] [Indexed: 11/18/2022]
Abstract
Electroacupuncture (EA) is reported to possess anti-inflammatory properties and has beneficial effects on acute respiratory distress syndrome (ARDS). However, the underlying mechanisms of the effects of EA on ARDS remain unclear. This study aims to investigate the protective effect of EA on LPS-induced ARDS. In this study, Sprague-Dawley male rats were treated with EA at Hegu (LI4) for 45 minutes before LPS instillation (0.4 mg/kg, 100 ul). H&E staining, wet-to-dry weight (W/D) ratio, PaO2, and protein content in BALF were employed to determine the function of lung tissues. Inflammatory cytokines in serum and BALF were detected by enzyme-linked immunoassay assay (ELISA). The levels of oxidative stress markers were detected to determine the oxidative stress status. Cell apoptosis was observed by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining and western blot. Here, we found that EA pretreatment effectively alleviated lung pathological damage. Moreover, EA suppressed the oxidative stress damage by upregulating glutathione and superoxide dismutase and downregulating malondialdehyde. EA pretreatment also regulated apoptosis-related proteins, such as Bax and Bcl-2. We found that peroxisome proliferators-activated receptors γ (PPARγ) play a critical role during ARDS, EA up-regulated the expression of PPARγ, which inhibited the activation of nuclear factor-kappa B (NF-κB) and decreased the inflammatory cytokines (interleukin-1β, interleukin-6, and tumor necrosis factor-α). When rats were treated with GW9662, a selective PPARγ antagonist, these effects of EA were reversed. Our study demonstrated that EA pretreatment had a beneficial effect on LPS-induced ARDS in rats by anti-inflammatory, antioxidative, and antiapoptotic properties which was regulated via PPARγ/NF-κB signaling pathway.
Collapse
|
29
|
d'Alessandro M, Bergantini L, Refini RM, Cameli P, Perillo F, Landi C, Icorne F, Perrone A, Sestini P, Bonella F, Bargagli E. Adiponectin and leptin levels in idiopathic pulmonary fibrosis: A new method for BAL and serum assessment. Immunobiology 2020; 225:151997. [PMID: 32962817 DOI: 10.1016/j.imbio.2020.151997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/06/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023]
Abstract
Adipokines (APN) are mainly secreted by adipocytes, macrophages and various other cells, along with their role in the regulation and mediation of inflammatory responses. APN is almost exclusively synthesized by adipocytes and regulated by peroxisome proliferator-activated receptor γ (PPARγ) that is involved in the epithelial-mesenchymal transition, linked lung fibrosis. Leptin is involved in acute lung injury with a role in lung fibrogenesis. Little is known about the relationship between APN/leptin and idiopathic pulmonary fibrosis (IPF) and the few studies available in the literature used ELISA to detect these lipid mediators. Our study is also the first to measure adipokines by the new multiplex assay and for the first time were performed in bronchoalveolar lavage (BAL) from IPF patients. This preliminary study suggests that APN levels in serum could be useful for predicting the prognosis of IPF, as they are inversely correlated with DLco percentages and BMI. Moreover, this first analysis of APN in BAL from IPF patients by a new method demonstrated an inverse correlation between these levels and BMI values and a direct correlation with eosinophil percentages, both of which are negative prognostic factors of IPF.
Collapse
Affiliation(s)
- Miriana d'Alessandro
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy.
| | - Laura Bergantini
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Rosa Metella Refini
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Paolo Cameli
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Felice Perillo
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Claudia Landi
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy; Functional proteomics lab, Department of life sciences, University of Siena, Italy
| | - Fiorella Icorne
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Anna Perrone
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Piersante Sestini
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Francesco Bonella
- Center for Interstitial and Rare Lung Disease, Department of Pulmonology, Ruhrlandklinik University Hospital, Essen, Germany
| | - Elena Bargagli
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| |
Collapse
|
30
|
Rebello CJ, Kirwan JP, Greenway FL. Obesity, the most common comorbidity in SARS-CoV-2: is leptin the link? Int J Obes (Lond) 2020; 44:1810-1817. [PMID: 32647360 PMCID: PMC7347260 DOI: 10.1038/s41366-020-0640-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/29/2020] [Accepted: 07/03/2020] [Indexed: 12/13/2022]
Abstract
Overweight and obesity are major risk factors for diabetes, cardiovascular disease, and lung disease. These diseases are the most commonly reported health conditions that predispose individuals with SARS-CoV-2 infection to require hospitalization including intensive care unit admissions. The innate immune response is the host’s first line of defense against a human coronavirus infection. However, most coronaviruses are armed with one strategy or another to overcome host antiviral defense, and the pathogenicity of the virus is related to its capacity to suppress host immunity. The multifaceted nature of obesity including its effects on immunity can fundamentally alter the pathogenesis of acute respiratory distress syndrome and pneumonia, which are the major causes of death due to SARS-CoV-2 infection. Elevated circulating leptin concentrations are a hallmark of obesity, which is associated with a leptin-resistant state. Leptin is secreted by adipocytes in proportion to body fat and regulates appetite and metabolism through signaling in the hypothalamus. However, leptin also signals through the Jak/STAT and Akt pathways, among others, to modulate T cell number and function. Thus, leptin connects metabolism with the immune response. Therefore, it seems appropriate that its dysregulation would have serious consequences during an infection. We propose that leptin may be the link between obesity and its high prevalence as a comorbidity of the SARS-CoV-2 infection. In this article, we present a synthesis of the mechanisms underpinning susceptibility to respiratory viral infections and the contribution of the immunomodulatory effects of obesity to the outcome.
Collapse
Affiliation(s)
- Candida J Rebello
- Pennington Biomedical Research Center, 6400, Perkins Road, Baton Rouge, LA 70808, USA
| | - John P Kirwan
- Pennington Biomedical Research Center, 6400, Perkins Road, Baton Rouge, LA 70808, USA
| | - Frank L Greenway
- Pennington Biomedical Research Center, 6400, Perkins Road, Baton Rouge, LA 70808, USA.
| |
Collapse
|
31
|
Dinger K, Koningsbruggen-Rietschel SV, Dötsch J, Alejandre Alcazar MA. Identification of Critical Windows of Metabolic Programming of Metabolism and Lung Function in Male Offspring of Obese Dams. Clin Transl Sci 2020; 13:1065-1070. [PMID: 32598577 PMCID: PMC7719392 DOI: 10.1111/cts.12811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/14/2020] [Indexed: 12/02/2022] Open
Abstract
Perinatal nutritional determinants known as metabolic programming could be either detrimental or protective. Maternal obesity in the perinatal period determines susceptibility for diseases, such as obesity, metabolic disorders, and lung disease. Although this adverse metabolic programming is well‐recognized, the critical developmental window for susceptibility risk remains elusive. Thus, we aimed to define the vulnerable window for impaired lung function after maternal obesity; and to test if dietary intervention protects. First, we studied the impact of high‐fat diet (HFD)‐induced maternal obesity during intrauterine (HFDiu), postnatal (HFDpost), or perinatal (i.e., intrauterine and postnatal (HFDperi) phase on body weight, white adipose tissue (WAT), glucose tolerance, and airway resistance. Although HFDiu, HFDpost, and HFDperi induced overweight in the offspring, only HFDperi and HFDiu led to increased WAT in the offspring early in life. This early‐onset adiposity was linked to impaired glucose tolerance in HFDperi‐offspring. Interestingly, these metabolic findings in HFDperi‐offspring, but not in HFDiu‐offspring and HFDpost‐offspring, were linked to persistent adiposity and increased airway resistance later in life. Second, we tested if the withdrawal of a HFD immediately after conception protects from early‐onset metabolic changes by maternal obesity. Indeed, we found a protection from early‐onset overweight, but not from impaired glucose tolerance and increased airway resistance. Our study identified critical windows for metabolic programming of susceptibility to impaired lung function, highlighting thereby windows of opportunity for prevention.
Collapse
Affiliation(s)
- Katharina Dinger
- Translational Experimental Pediatrics - Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Silke V Koningsbruggen-Rietschel
- Pediatric Pulmonology, Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Miguel A Alejandre Alcazar
- Translational Experimental Pediatrics - Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
32
|
Abstract
For infectious-disease outbreaks, clinical solutions typically focus on efficient pathogen destruction. However, the COVID-19 pandemic provides a reminder that infectious diseases are complex, multisystem conditions, and a holistic understanding will be necessary to maximize survival. For COVID-19 and all other infectious diseases, metabolic processes are intimately connected to the mechanisms of disease pathogenesis and the resulting pathology and pathophysiology, as well as the host defence response to the infection. Here, I examine the relationship between metabolism and COVID-19. I discuss why preexisting metabolic abnormalities, such as type 2 diabetes and hypertension, may be important risk factors for severe and critical cases of infection, highlighting parallels between the pathophysiology of these metabolic abnormalities and the disease course of COVID-19. I also discuss how metabolism at the cellular, tissue and organ levels might be harnessed to promote defence against the infection, with a focus on disease-tolerance mechanisms, and speculate on the long-term metabolic consequences for survivors of COVID-19.
Collapse
Affiliation(s)
- Janelle S Ayres
- Molecular and Systems Physiology Laboratory, Gene Expression Laboratory, NOMIS Center for Immunology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
33
|
d'Alessandro M, Bergantini L, Cameli P, Lanzarone N, Perillo F, Perrone A, Bargagli E. BAL and serum multiplex lipid profiling in idiopathic pulmonary fibrosis and fibrotic hypersensitivity pneumonitis. Life Sci 2020; 256:117995. [PMID: 32574666 DOI: 10.1016/j.lfs.2020.117995] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Differential diagnosis between IPF and fibrotic HP (fHP) can be challenging: these two ILDs share many common features but call for different therapeutic approaches. In the present study, differential lipid mediator profiles were analysed by a new method in BAL and serum from HP and IPF patients. MATERIALS AND METHODS 76 patients were enrolled retrospectively in the study. Median age (IQR) was 67 years (51-74); 63% were males, 30 had fHP and 46 had IPF. Serum and BAL samples were collected at initial diagnosis. For quantification of serum and BAL lipid mediators was used bead-based multiplex LEGENDPlex™ analysis (Biolegend). RESULTS Serum Apo A1 levels were significantly higher in IPF than fHP patients (p = 0.314); indeed, serum levels of CCL2 and Apo C3 were lower in HP than in IPF patients (p = 0.013 and p = 0.041, respectively). BAL concentrations of Apo A1, adipsin, Apo C3 and APN were significantly lower in IPF than in fHP patients (p < 0.0001, p < 0.0001, p = 0.007 and p = 0.023, respectively). In the logistic regression, IPF was tested as dependent variable. Serum levels of Apo A1, CCL2 and Apo C3 were tested as independent variables and ROC curve analysis of model performance showed AUC 93% (p < 0.0001); on the other hand, BAL concentrations of Apo A1, adipsin, Apo C3 and APN showed AUC 81% (p < 0.0001). DISCUSSION Lipid biomarkers evaluated in BAL in our study confirm the hypothesis that fHP and IPF have different lung fibrosis phenotypes. The former is a post-inflammatory cell-regulated ILD and the second is more related to tissue remodeling and repair.
Collapse
Affiliation(s)
- Miriana d'Alessandro
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy.
| | - Laura Bergantini
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Paolo Cameli
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Nicola Lanzarone
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Felice Perillo
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Anna Perrone
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Elena Bargagli
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| |
Collapse
|
34
|
Yin B, Jiang H, Liu X, Guo SW. Enriched Environment Decelerates the Development of Endometriosis in Mouse. Reprod Sci 2020; 27:1423-1435. [PMID: 32318984 DOI: 10.1007/s43032-019-00117-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/21/2019] [Indexed: 12/16/2022]
Abstract
We tested the hypothesis that enriched environment (EE), consisting of enlarged space, and increased physical activity and social interactions, hinders the development of endometriosis through attenuated adrenergic signaling, enhanced autophagy, and reduced leptin levels. Two mouse experiments were performed. In Experiment 1, 40 female Balb/C mice were randomly divided into four equal-sized groups, the SE (standard environment), EE, p-EE (EE instituted after endometriosis induction), and the d-EE (SE housing but received uterine fragments from EE donors) groups. Housing intervention was initiated 3 weeks before the induction of endometriosis and continued for 3 weeks after induction. In Experiment 2, 20 female mice were randomly divided into SE and EE groups, and the plasma leptin levels were measured. We measured lesion weight and hotplate latency and performed Masson trichrome staining as well as immunohistochemistry analysis of β2 adrenergic receptor (ADRB2), dopamine receptor D2 (DRD2), vascular endothelial growth factor (VEGF), and microtubule-associated protein light chain 3 (LC3). We found that EE reduced the lesion weight by 40.8% as compared with SE mice, but the reduction in p-EE and d-EE mice did not reach statistical significance. EE significantly reduced staining levels of ADRB2 and VEGF as well as the extent of lesional fibrosis but increased staining levels of LC3 and DRD2 in lesions as compared with the SE group. EE mice had reduced plasma leptin levels as compared with SE mice. Thus, EE decelerates the development of endometriosis and fibrogenesis and improved generalized hyperalgesia, possibly through increased DRD2 expression but decreased expression of ADRB2 and VEGF as well as enhanced autophagy and reduced leptin level.
Collapse
Affiliation(s)
- Bo Yin
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, 200011, China
| | - Hongyuan Jiang
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, 200011, China
| | - Xishi Liu
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, 200011, China. .,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China.
| | - Sun-Wei Guo
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, 200011, China. .,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China.
| |
Collapse
|
35
|
Kim JS, Anderson MR, Podolanczuk AJ, Kawut SM, Allison MA, Raghu G, Hinckley-Stuckovsky K, Hoffman EA, Tracy RP, Barr RG, Lederer DJ, Giles JT. Associations of Serum Adipokines With Subclinical Interstitial Lung Disease Among Community-Dwelling Adults: The Multi-Ethnic Study of Atherosclerosis (MESA). Chest 2020; 157:580-589. [PMID: 31678306 PMCID: PMC7078588 DOI: 10.1016/j.chest.2019.10.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/03/2019] [Accepted: 10/06/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Adipokines have inflammatory and fibrotic properties that may be critical in interstitial lung disease (ILD). We examined associations of serum adipokine levels with CT imaging-based measures of subclinical ILD and lung function among community-dwelling adults. METHODS A subset of the original Multi-Ethnic Study of Atherosclerosis cohort (n = 1,968) had adiponectin, leptin, and resistin measured during follow-up visits (2002-2005). We used regression models to examine associations of adiponectin, leptin, and resistin levels with (1) high-attenuation areas (HAAs) from CT scans (2004-2005, n = 1,144), (2) interstitial lung abnormalities (ILAs) from CT scans (2010-2012, n = 872), and (3) FVC from spirometry (2004-2006, n = 1,446). We used -(1/HAA2), which we denoted with H, to model HAA as our outcome to meet model assumptions. RESULTS Higher adiponectin was associated with lower HAA on CT imaging among adults with a BMI ≥ 25 kg/m2 (P for BMI interaction = .07). Leptin was more strongly associated with ILA among never smokers compared with ever smokers (P for smoking interaction = .004). For every 1-SD increment of log-transformed leptin, the percent predicted FVC was 3.8% lower (95% CI, -5.0 to -2.5). Higher serum resistin levels were associated with greater HAA on CT in a fully adjusted model. For every 1-SD increment of log-transformed resistin there was an increase in H of 14.8 (95% CI, 3.4-26.3). CONCLUSIONS Higher adiponectin levels were associated with lower HAA on CT imaging among adults with a higher BMI. Higher leptin and resistin levels were associated with lower FVC and greater HAA, respectively.
Collapse
Affiliation(s)
- John S Kim
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA.
| | | | - Anna J Podolanczuk
- Department of Medicine, Columbia University Medical Center, New York, NY
| | - Steven M Kawut
- Department of Medicine and the Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Matthew A Allison
- Department of Family and Preventative Medicine, University of California San Diego, San Diego, CA
| | - Ganesh Raghu
- Department of Medicine, University of Washington, Seattle, WA
| | | | - Eric A Hoffman
- Departments of Radiology, Medicine, and Biomedical Engineering, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Russell P Tracy
- Departments of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT
| | - R Graham Barr
- Department of Medicine, Columbia University Medical Center, New York, NY; Department of Epidemiology, Mailman School of Public Health, Columbia University Medical Center, New York, NY
| | - David J Lederer
- Department of Medicine, Columbia University Medical Center, New York, NY; Department of Epidemiology, Mailman School of Public Health, Columbia University Medical Center, New York, NY
| | - Jon T Giles
- Department of Medicine, Columbia University Medical Center, New York, NY
| |
Collapse
|
36
|
Abstract
Leptin is a hormone that plays a major role as mediator of long-term regulation of energy balance, suppressing food intake, and stimulating weight loss. More recently, important physiological roles other than controlling appetite and energy expenditure have been suggested for leptin, including neuroendocrine, reparative, reproductive, and immune functions. These emerging peripheral roles let hypothesize that leptin can modulate also cancer progression. Indeed, many studies have demonstrated that elevated chronic serum concentrations of leptin, frequently seen in obese subjects, represent a stimulatory signal for tumor growth. Current knowledge indicates that also different non-tumoral cells resident in tumor microenvironment may respond to leptin creating a favorable soil for cancer cells. In addition, leptin is produced also within the tumor microenvironment creating the possibility for paracrine and autocrine action. In this review, we describe the main mechanisms that regulate peripheral leptin availability and how leptin can shape tumor microenvironment.
Collapse
|
37
|
Khaing P, Pandit P, Awsare B, Summer R. Pulmonary Circulation in Obesity, Diabetes, and Metabolic Syndrome. Compr Physiol 2019; 10:297-316. [DOI: 10.1002/cphy.c190018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
38
|
Anderson MR, Udupa JK, Edwin E, Diamond JM, Singer JP, Kukreja J, Hays SR, Greenland JR, Ferrante A, Lippel M, Blue T, McBurnie A, Oyster M, Kalman L, Rushefski M, Wu C, Pednekar G, Liu W, Arcasoy S, Sonett J, D'Ovidio F, Bacchetta M, Newell JD, Torigian D, Cantu E, Farber DL, Giles JT, Tong Y, Palmer S, Ware LB, Hancock WW, Christie JD, Lederer DJ. Adipose tissue quantification and primary graft dysfunction after lung transplantation: The Lung Transplant Body Composition study. J Heart Lung Transplant 2019; 38:1246-1256. [PMID: 31474492 PMCID: PMC6883162 DOI: 10.1016/j.healun.2019.08.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 07/30/2019] [Accepted: 08/05/2019] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Obesity is associated with an increased risk of primary graft dysfunction (PGD) after lung transplantation. The contribution of specific adipose tissue depots is unknown. METHODS We performed a prospective cohort study of adult lung transplant recipients at 4 U.S. transplant centers. We measured cross-sectional areas of subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) on chest and abdominal computed tomography (CT) scans and indexed each measurement to height.2 We used logistic regression to examine the associations of adipose indices and adipose classes with grade 3 PGD at 48 or 72 hours, and Cox proportional hazards models to examine survival. We used latent class analyses to identify the patterns of adipose distribution. We examined the associations of adipose indices with plasma biomarkers of obesity and PGD. RESULTS A total of 262 and 117 subjects had available chest CT scans and underwent protocol abdominal CT scans, respectively. In the adjusted models, a greater abdominal SAT index was associated with an increased risk of PGD (odds ratio 1.9, 95% CI 1.02-3.4, p = 0.04) but not with survival time. VAT indices were not associated with PGD risk or survival time. A greater abdominal SAT index correlated with greater pre- and post-transplant leptin (r = 0.61, p < 0.001, and r = 0.44, p < 0.001), pre-transplant IL-1RA (r = 0.25, p = 0.04), and post-transplant ICAM-1 (r = 0.25, p = 0.04). We identified 3 latent patterns of adiposity. The class defined by high thoracic and abdominal SAT had the greatest risk of PGD. CONCLUSIONS Subcutaneous, but not visceral, adiposity is associated with an increased risk of PGD after lung transplantation.
Collapse
Affiliation(s)
- Michaela R Anderson
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Jayaram K Udupa
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ethan Edwin
- Columbia Institute of Human Nutrition, Columbia University Medical Center, New York, New York
| | - Joshua M Diamond
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jonathan P Singer
- Department of Medicine University of California at San Francisco, San Francisco, California
| | - Jasleen Kukreja
- Department of Surgery, University of California at San Francisco, San Francisco, California
| | - Steven R Hays
- Department of Medicine University of California at San Francisco, San Francisco, California
| | - John R Greenland
- Department of Medicine University of California at San Francisco, San Francisco, California
| | - Anthony Ferrante
- Columbia Institute of Human Nutrition, Columbia University Medical Center, New York, New York
| | - Matthew Lippel
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Tatiana Blue
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Amika McBurnie
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Michelle Oyster
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Laurel Kalman
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Melanie Rushefski
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Caiyun Wu
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gargi Pednekar
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Wen Liu
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Selim Arcasoy
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Joshua Sonett
- Department of Surgery, Columbia University Medical Center, New York, New York
| | - Frank D'Ovidio
- Department of Surgery, Columbia University Medical Center, New York, New York
| | - Matthew Bacchetta
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - John D Newell
- Department of Radiology, University of Iowa, Iowa City, Iowa
| | - Drew Torigian
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edward Cantu
- Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Donna L Farber
- Department of Surgery, University of California at San Francisco, San Francisco, California; Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York; Department of Microbiology and Immunology, Columbia University Medical Center, New York, New York
| | - Jon T Giles
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Yubing Tong
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Scott Palmer
- Department of Medicine, Duke University & Duke Clinical Research Institute, Durham, North Carolina
| | - Lorraine B Ware
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Wayne W Hancock
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jason D Christie
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David J Lederer
- Department of Medicine, Columbia University Medical Center, New York, New York; Department of Epidemiology, Mailman School of Public Health, Columbia University Medical Center, New York, New York.
| |
Collapse
|
39
|
Enomoto N, Oyama Y, Yasui H, Karayama M, Hozumi H, Suzuki Y, Kono M, Furuhashi K, Fujisawa T, Inui N, Nakamura Y, Suda T. Analysis of serum adiponectin and leptin in patients with acute exacerbation of idiopathic pulmonary fibrosis. Sci Rep 2019; 9:10484. [PMID: 31324858 PMCID: PMC6642131 DOI: 10.1038/s41598-019-46990-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 07/09/2019] [Indexed: 01/19/2023] Open
Abstract
Weight loss progresses with the progression of idiopathic pulmonary fibrosis (IPF), and acute exacerbation of IPF (AE-IPF) frequently occurs in its advanced stage. Adiponectin and leptin are adipokines produced from adipose tissue, and are related to thinness and obesity, respectively. Additionally, these adipokines are implicated in the regulation of inflammation and fibrosis centering on peroxisome proliferator-activated receptor γ (PPARγ). However, the relationship between adiponectin/leptin and AE-IPF remains poorly known. We conducted this study to evaluate levels of serum adiponectin/leptin, and to elucidate the clinical importance of adiponectin and leptin in patients with AE-IPF. Thirty-two patients (39 episodes) who were diagnosed with AE-IPF at our hospital from 1997 to 2016 were retrospectively studied. Serum adiponectin and leptin concentrations were measured with enzyme-linked immunosorbent assay. Patients with AE-IPF showed higher levels of serum adiponectin and leptin than those at initial diagnosis of IPF (p = 0.007 and p = 0.027, respectively). Serum adiponectin/leptin (A/L) ratio was negatively correlated with body mass index at AE-IPF (r = -0.456, p = 0.003) and PaO2 before AE-IPF (r = -0.498, p = 0.034), and positively correlated with C-reactive protein at AE-IPF (r = 0.316, p = 0.049). Patients with higher A/L ratios had worse survival than those with lower A/L ratios (log-rank, p = 0.026). Further, in multivariate analysis, serum A/L ratio was a significant prognostic factor in patients with AE-IPF (HR 2.60, p = 0.042). In conclusion, the higher adiponectin/leptin ratio may be associated with a poor prognosis in patients with AE-IPF.
Collapse
Affiliation(s)
- Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan. .,Health Administration Center, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Yoshiyuki Oyama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hideki Yasui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masato Karayama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuzo Suzuki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masato Kono
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuki Furuhashi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.,Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yutaro Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
40
|
Michalska-Jakubus M, Sawicka K, Potembska E, Kowal M, Krasowska D. Clinical associations of serum leptin and leptin/adiponectin ratio in systemic sclerosis. Postepy Dermatol Alergol 2019; 36:325-338. [PMID: 31333350 PMCID: PMC6640022 DOI: 10.5114/ada.2018.75809] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 04/06/2018] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Leptin and adiponectin have recently received the attention of researchers as attractive biomarkers in systemic sclerosis (SSc) because of their role in the inflammatory process, vascular function and fibrosis. We hypothesized that leptin and adiponectin may be associated with disease activity and severity in patients with SSc. AIM To compare serum leptin, adiponectin and leptin/adiponectin levels in patients with SSc and healthy controls and to evaluate their possible relationship with frequently used laboratory markers and clinical findings. MATERIAL AND METHODS The study included 48 Caucasian female patients with SSc and 38 healthy controls. Serum concentrations of leptin and adiponectin were measured in patients and controls using commercially available ELISA Kits (Quantikine ELISA Kit R&D Systems, Minneapolis, MN, USA). The results were assessed by the Mann-Whitney U-test and Spearman's correlation test. RESULTS Leptin and adiponectin levels correlated with body mas index in SSc patients (r = 0.495, p = 0.000398 and r = -0.306; p = 0.0342) in contrast to healthy controls (p = 0.070 and p = 0.256, respectively), and, in SSc patients only, a strong negative correlation was observed between leptin and adiponectin serum levels (r = -0.314; p = 0.0312). Diffuse form of the disease (dcSSc) was associated with significantly lower serum adiponectin levels (8638.62 ±10382.62). Active disease was associated with significantly lower leptin concentration (13700.49 ±18293.32) and there was a significant negative correlation between leptin serum level and activity index score (r = -0.342; p = 0.0185). CONCLUSIONS The results of our study indicate that leptin levels might correlate with disease activity and subtype in SSc patients.
Collapse
Affiliation(s)
| | - Karolina Sawicka
- Department of Dermatology, Venereology and Pediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Emilia Potembska
- Department of Psychiatric Nursing, Medical University of Lublin, Lublin, Poland
| | - Małgorzata Kowal
- Department of Dermatology, Venereology and Pediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Dorota Krasowska
- Department of Dermatology, Venereology and Pediatric Dermatology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
41
|
Bruno A, Di Sano C, Lorusso F, Dino P, Russo D, Ballacchino A, Gallina S, Modica DM, Chiappara G, Simon HU, Pace E. Notch-1 decreased expression contributes to leptin receptor downregulation in nasal epithelium from allergic turbinates. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1642-1650. [PMID: 30951821 DOI: 10.1016/j.bbadis.2019.03.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Allergic rhinitis is characterized by a remodeling of nasal epithelium. Since the Notch and TGF-β signaling pathways are known to be involved in cell differentiation and remodeling processes and leptin adipokine has already been identified as a marker for homeostasis in human bronchial and nasal epithelial cells of asthmatics, roles played by these pathways have been investigated for chronic allergic rhinitis. METHODS The leptin/leptin receptor expression has been investigated in a study with 40 biopsies from allergic (AR, n = 18) and non-allergic (C, n = 22) inferior turbinates, using immunohistochemistry, immunofluorescence staining and RT-PCR. In addition, extracts from in vitro samples prepared from primary cells of inferior turbinates as well as in vitro cultured human nasal epithelial RPMI 2650 cells (ATCC-CCL-30) were also tested for leptin expression and activation of the Notch-1 pathway. RESULTS With regards to AR, in vivo expression levels of both leptin and its receptor significantly decreased in comparison to C. Furthermore, leptin receptor mRNA was significantly reduced in AR as compared to C. Immunofluorescence showed an apparent co-expression of leptin receptor with Notch-1, which was not seen with TGF-β. In vitro, in primary turbinate epithelial cells, the expression of leptin receptor and Notch-1 significantly decreased in AR as compared to C. Moreover, in RPMI 2650 cells, leptin receptor expression was shown to be induced by Notch-1 ligand signaling. CONCLUSION Thus, both the leptin and Notch-1 pathways appear to represent markers for epithelial homeostasis in allergic rhinitis.
Collapse
Affiliation(s)
- Andreina Bruno
- CNR, Institute of Biomedicine and Molecular Immunology, Palermo, Italy
| | - Caterina Di Sano
- CNR, Institute of Biomedicine and Molecular Immunology, Palermo, Italy
| | - Francesco Lorusso
- Otorhinolaryngology Section, Department of Experimental Biomedicine and Clinical Neurosciences, (BioNeC), University of Palermo, Italy
| | - Paola Dino
- CNR, Institute of Biomedicine and Molecular Immunology, Palermo, Italy
| | - Domenica Russo
- CNR, Institute of Biomedicine and Molecular Immunology, Palermo, Italy
| | | | - Salvatore Gallina
- Otorhinolaryngology Section, Department of Experimental Biomedicine and Clinical Neurosciences, (BioNeC), University of Palermo, Italy
| | - Domenico Michele Modica
- Otorhinolaryngology Section, Department of Experimental Biomedicine and Clinical Neurosciences, (BioNeC), University of Palermo, Italy
| | | | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Elisabetta Pace
- CNR, Institute of Biomedicine and Molecular Immunology, Palermo, Italy
| |
Collapse
|
42
|
Alipoor E, Mohammad Hosseinzadeh F, Hosseinzadeh-Attar MJ. Adipokines in critical illness: A review of the evidence and knowledge gaps. Biomed Pharmacother 2018; 108:1739-1750. [PMID: 30372877 DOI: 10.1016/j.biopha.2018.09.165] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/27/2018] [Accepted: 09/28/2018] [Indexed: 12/28/2022] Open
Abstract
Adipose tissue products or adipokines play a major role in chronic endocrine and metabolic disorders; however, little is known about critical conditions. In this article, the experimental and clinical evidence of alterations of adipokines, adiponectin, leptin, resistin, visfatin, asymmetric dimethylarginine (ADMA), and ghrelin in critical illness, their potential metabolic, diagnostic, and prognostic value, and the gaps in the field have been reviewed. The results showed considerable changes in the concentration of the adipokines; while the impact of adipokines on metabolic disorders such as insulin resistance and inflammation has not been well documented in critically ill patients. There is no consensus about the circulatory and functional changes of leptin and adiponectin. However, it seems that lower concentrations of adiponectin at admission with gradual consequent increase might be a useful pattern in determining better outcomes of critical illness. Some evidence has suggested the adverse effects of elevated resistin concentration, potential prognostic importance of visfatin, and therapeutic value of ghrelin. High ADMA levels and low arginine:ADMA ratio were also proposed as predictors of ICU mortality and morbidities. However, there is no consensus on these findings. Although primary data indicated the role of adipokines in critical illness, further studies are required to clarify whether the reason of these changes is pathophysiological or compensatory. The relationship of pathophysiological background, disease severity, baseline nutritional status and nutrition support during hospitalization, and variations in body fat percentage and distribution with adipokines, as well as the potential prognostic or therapeutic role of these peptides should be further investigated in critically ill patients.
Collapse
Affiliation(s)
- Elham Alipoor
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran; Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mohammad Hosseinzadeh
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Hosseinzadeh-Attar
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran; Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
43
|
Hamanaka RB, Nigdelioglu R, Meliton AY, Tian Y, Witt LJ, O’Leary E, Sun KA, Woods PS, Wu D, Ansbro B, Ard S, Rohde JM, Dulin NO, Guzy RD, Mutlu GM. Inhibition of Phosphoglycerate Dehydrogenase Attenuates Bleomycin-induced Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2018; 58:585-593. [PMID: 29019702 PMCID: PMC5946329 DOI: 10.1165/rcmb.2017-0186oc] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 09/14/2017] [Indexed: 12/13/2022] Open
Abstract
Organ fibrosis, including idiopathic pulmonary fibrosis, is associated with significant morbidity and mortality. Because currently available therapies have limited effect, there is a need to better understand the mechanisms by which organ fibrosis occurs. We have recently reported that transforming growth factor (TGF)-β, a key cytokine that promotes fibrogenesis, induces the expression of the enzymes of the de novo serine and glycine synthesis pathway in human lung fibroblasts, and that phosphoglycerate dehydrogenase (PHGDH; the first and rate-limiting enzyme of the pathway) is required to promote collagen protein synthesis downstream of TGF-β. In this study, we investigated whether inhibition of de novo serine and glycine synthesis attenuates lung fibrosis in vivo. We found that TGF-β induces mRNA and protein expression of PHGDH in murine fibroblasts. Similarly, intratracheal administration of bleomycin resulted in increased expression of PHGDH in mouse lungs, localized to fibrotic regions. Using a newly developed small molecule inhibitor of PHGDH (NCT-503), we tested whether pharmacologic inhibition of PHGDH could inhibit fibrogenesis both in vitro and in vivo. Treatment of murine and human lung fibroblasts with NCT-503 decreased TGF-β-induced collagen protein synthesis. Mice treated with the PHGDH inhibitor beginning 7 days after intratracheal instillation of bleomycin had attenuation of lung fibrosis. These results indicate that the de novo serine and glycine synthesis pathway is necessary for TGF-β-induced collagen synthesis and bleomycin-induced pulmonary fibrosis. PHGDH and other enzymes in the de novo serine and glycine synthesis pathway may be a therapeutic target for treatment of fibrotic diseases, including idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Robert B. Hamanaka
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, Illinois; and
| | - Recep Nigdelioglu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, Illinois; and
| | - Angelo Y. Meliton
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, Illinois; and
| | - Yufeng Tian
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, Illinois; and
| | - Leah J. Witt
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, Illinois; and
| | - Erin O’Leary
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, Illinois; and
| | - Kaitlyn A. Sun
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, Illinois; and
| | - Parker S. Woods
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, Illinois; and
| | - David Wu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, Illinois; and
| | - Brandon Ansbro
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, Illinois; and
| | - Shawn Ard
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, Illinois; and
| | - Jason M. Rohde
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland
| | - Nickolai O. Dulin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, Illinois; and
| | - Robert D. Guzy
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, Illinois; and
| | - Gökhan M. Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, Illinois; and
| |
Collapse
|
44
|
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, Dallas, TX
| |
Collapse
|
45
|
Leptin-induced inflammation by activating IL-6 expression contributes to the fibrosis and hypertrophy of ligamentum flavum in lumbar spinal canal stenosis. Biosci Rep 2018; 38:BSR20171214. [PMID: 29436483 PMCID: PMC5874260 DOI: 10.1042/bsr20171214] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/15/2017] [Accepted: 12/16/2017] [Indexed: 01/07/2023] Open
Abstract
The ongoing chronic inflammation and subsequent fibrosis play an important role in ligamentum flavum (LF) fibrosis and hypertrophy in patients with lumbar spinal canal stenosis (LSCS). Leptin is a chronic inflammatory mediator and involved in the fibrotic process in multiple organ systems. The present study aimed to investigate the role of leptin in LF fibrosis and its related regulatory mechanisms. The LF specimens were obtained during the surgery from 12 patients with LSCS (LSCS group) and 12 control patients with lumbar disc herniation (LDH) group. The morphologic changes and fibrosis score of LF were assessed by Hematoxylin and eosin (H&E) and Masson’s trichrome staining respectively. The location and expression of leptin in LF tissues were determined. Then, the LF cells were cultured and exposed to recombinant human leptin (rhleptin). Collagen I and III were used as fibrosis markers and IL-6 was used as the inflammatory factor. As a result, the LF thickness and fibrosis score in the LSCS group were significantly higher than those in the LDH group (P<0.05). Leptin was detected in the hypertrophied LF and its expression was substantially increased in the LSCS group and positively correlated with LF thickness and fibrosis score (P<0.05). Moreover, our in vitro experiments revealed that rhleptin treated LF cells elevated the expression of collagen I and III. Finally, leptin administration induced IL-6 expression via nuclear factor-κB (NF-κB) pathway in LF cell (P<0.05). Our study demonstrated novel molecular events for leptin-induced inflammation in LF tissue by promoting IL-6 expression and thus might have potential implications for clarifying the mechanism underlying LF fibrosis and hypertrophy.
Collapse
|
46
|
Gui X, Chen H, Cai H, Sun L, Gu L. Leptin promotes pulmonary fibrosis development by inhibiting autophagy via PI3K/Akt/mTOR pathway. Biochem Biophys Res Commun 2018. [PMID: 29524411 DOI: 10.1016/j.bbrc.2018.03.039] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Leptin, a protein-related product of the obesity gene, plays an important role in the pathogenesis of fibrotic diseases including pulmonary fibrosis. As a highly conservative process, autophagy regulates various biological functions. Otherwise, insufficient autophagy has been described in alveolar epithelial cells (AEC) to cope with the progression of pulmonary fibrosis. Hence, this study is to investigate the effects of leptin on fibrosis in TGF-β1 induced epithelial mesenchymal transition (EMT) and the potential roles of autophagy in this processes. Our results showed that the elevated leptin level in serum correlated with the severity of lung fibrosis and leptin significantly promoted the EMT in A549 cells as evidenced by promoting collagen I and α-SMA production. Additionally, treatment with leptin decreased autophagosome formation, inhibited the lipidation of LC3I to LC3II, and up-regulated the expression of p62 via activating PI3K/Akt/mTOR pathway, which is indicative of inhibition of autophagy by leptin. Finally, rapmycin pretreatment reversed the pro-fibrogenic effects of leptin. Taken together, our study suggested that leptin accelerated the EMT of A549 cells through inhibiting autophagy via PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Xianhua Gui
- Department of Respiratory Medicine, The Affiliated Nanjing Drum Tower clinical medical college of Nanjing Medical University, 210008, Nanjing, China; Department of Respiratory Medicine, The Affiliated Drum Tower Hospital, Nanjing University Medical School, 210008, Nanjing, China
| | - Hongwei Chen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, 210008, Nanjing, China
| | - Hourong Cai
- Department of Respiratory Medicine, The Affiliated Nanjing Drum Tower clinical medical college of Nanjing Medical University, 210008, Nanjing, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Nanjing Drum Tower Clinical Medical College of Nanjing Medical University, 210008, Nanjing, China.
| | - Luo Gu
- Department of Physiology of Nanjing Medical University, 211166, Nanjing, China.
| |
Collapse
|
47
|
Xie X, Li S, Zhu Y, Liu L, Ke R, Wang J, Yan X, Yang L, Gao L, Zang W, Li M. Egr-1 mediates leptin-induced PPARγ reduction and proliferation of pulmonary artery smooth muscle cells. Mol Biol Cell 2017; 29:356-362. [PMID: 29212876 PMCID: PMC5996952 DOI: 10.1091/mbc.e17-03-0141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 11/08/2017] [Accepted: 11/27/2017] [Indexed: 01/14/2023] Open
Abstract
Loss of peroxisome proliferator-activated receptor γ (PPARγ) has been found to contribute to pulmonary artery smooth muscle cell (PASMC) proliferation and pulmonary arterial remodeling therefore the development of pulmonary hypertension (PH). Yet, the molecular mechanisms underlying PPARγ reduction in PASMC remain poorly understood. Here, we demonstrated that leptin dose- and time-dependently inducued PPARγ down-regulation and proliferation of primary cultured rat PASMC, this was accompanied with the activation of extracellular regulated kinase1/2 (ERK1/2) signaling pathway and subsequent induction of early growth response-1 (Egr-1) expression. The presence of MEK inhibitors U0126 or PD98059, or prior silencing Egr-1 with small interfering RNA suppressed leptin-induced PPARγ reduction. In addition, activation of PPARγ by pioglitazone or targeting ERK1/2/Egr-1 suppressed leptin-induced PASMC proliferation. Taken together, our study indicates that ERK1/2 signaling pathway-mediated leptin-induced PPARγ reduction and PASMC proliferation through up-regulation of Egr-1 and suggests that targeting leptin/ERK1/2/Egr-1 pathway might have potential value in ameliorating vascular remodeling and benefit PH.
Collapse
Affiliation(s)
- Xinming Xie
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yanting Zhu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Lu Liu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Rui Ke
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xin Yan
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Lan Yang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Li Gao
- Division of Allergy and Clinical Immunology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Weijin Zang
- Department of Pharmacology, School of Basic Medical Sciences, Xian Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
48
|
Bouros E, Filidou E, Arvanitidis K, Mikroulis D, Steiropoulos P, Bamias G, Bouros D, Kolios G. Lung fibrosis-associated soluble mediators and bronchoalveolar lavage from idiopathic pulmonary fibrosis patients promote the expression of fibrogenic factors in subepithelial lung myofibroblasts. Pulm Pharmacol Ther 2017; 46:78-87. [PMID: 28865842 DOI: 10.1016/j.pupt.2017.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 08/29/2017] [Indexed: 01/13/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by infiltration of inflammatory cells, excessive collagen production and accumulation of myofibroblasts. We explored the possible role of subepithelial lung myofibroblasts (SELMs) in the development of fibrosis in IPF. SELMs, isolated from surgical specimens of healthy lung tissue, were cultured with pro-inflammatory factors or bronchoalveolar lavage fluid (BALF) from patients with IPF or idiopathic non-specific interstitial pneumonia (iNSIP) and their fibrotic activity was assessed. Stimulation of SELMs with pro-inflammatory factors induced a significant increase of Tissue Factor (TF) and Tumor necrosis factor-Like cytokine 1 A (TL1A) expression and collagen production in culture supernatants. Stimulation with BALF from IPF patients with mild to moderate, but not severe disease, and from iNSIP patients induced a significant increase of TF expression. BALF from all IPF patients induced a significant increase of TL1A expression and collagen production, while BALF from iNSIP patients induced a significant increase of TL1A, but not of collagen production. Interestingly, TGF-β1 and BALF from all IPF, but not iNSIP patients, induced a significant increase in SELMs migration. In conclusion, BALF from IPF patients induces fibrotic activity in lung myofibroblasts, similar to mediators associated with lung fibrosis, indicating a key role of SELMs in IPF.
Collapse
Affiliation(s)
- Evangelos Bouros
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.
| | - Eirini Filidou
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.
| | - Konstantinos Arvanitidis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.
| | - Dimitrios Mikroulis
- Thoracic Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.
| | - Paschalis Steiropoulos
- Pneumonology Department, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.
| | - George Bamias
- Gastroenterology Department of Laiko Hospital, Medical School, National and Kapodistrian University, Athens, Greece.
| | - Demosthenes Bouros
- Interstitial Lung Disease Unit of 1st Department of Pneumonology, Medical School, National and Kapodistrian University, Athens, Greece.
| | - George Kolios
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.
| |
Collapse
|
49
|
Ubags NDJ, Burg E, Antkowiak M, Wallace AM, Dilli E, Bement J, Wargo MJ, Poynter ME, Wouters EFM, Suratt BT. A Comparative Study of Lung Host Defense in Murine Obesity Models. Insights into Neutrophil Function. Am J Respir Cell Mol Biol 2017; 55:188-200. [PMID: 27128821 DOI: 10.1165/rcmb.2016-0042oc] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
We have shown that obesity-associated attenuation of murine acute lung injury is driven, in part, by blunted neutrophil chemotaxis, yet differences were noted between the two models of obesity studied. We hypothesized that obesity-associated impairment of multiple neutrophil functions contributes to increased risk for respiratory infection but that such impairments may vary between murine models of obesity. We examined the most commonly used murine obesity models (diet-induced obesity, db/db, CPE(fat/fat), and ob/ob) using a Klebsiella pneumoniae pneumonia model and LPS-induced pneumonitis. Marrow-derived neutrophils from uninjured lean and obese mice were examined for in vitro functional responses. All obesity models showed impaired clearance of K. pneumoniae, but in differing temporal patterns. Failure to contain infection in obese mice was seen in the db/db model at both 24 and 48 hours, yet this defect was only evident at 24 hours in CPE(fat/fat) and ob/ob models, and at 48 hours in diet-induced obesity. LPS-induced airspace neutrophilia was decreased in all models, and associated with blood neutropenia in the ob/ob model but with leukocytosis in the others. Obese mouse neutrophils from all models demonstrated impaired chemotaxis, whereas neutrophil granulocyte colony-stimulating factor-mediated survival, LPS-induced cytokine transcription, and mitogen-activated protein kinase and signal transducer and activator of transcription 3 activation in response to LPS and granulocyte colony-stimulating factor, respectively, were variably impaired across the four models. Obesity-associated impairment of host response to lung infection is characterized by defects in neutrophil recruitment and survival. However, critical differences exist between commonly used mouse models of obesity and may reflect variable penetrance of elements of the metabolic syndrome, as well as other factors.
Collapse
Affiliation(s)
- Niki D J Ubags
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, the Netherlands; and.,Departments of 2 Medicine and
| | | | | | | | | | | | - Matthew J Wargo
- 3 Microbiology and Molecular Genetics, University of Vermont College of Medicine, Burlington, Vermont
| | | | - Emiel F M Wouters
- 1 Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, the Netherlands; and
| | | |
Collapse
|
50
|
Nigdelioglu R, Hamanaka RB, Meliton AY, O'Leary E, Witt LJ, Cho T, Sun K, Bonham C, Wu D, Woods PS, Husain AN, Wolfgeher D, Dulin NO, Chandel NS, Mutlu GM. Transforming Growth Factor (TGF)-β Promotes de Novo Serine Synthesis for Collagen Production. J Biol Chem 2016; 291:27239-27251. [PMID: 27836973 PMCID: PMC5207151 DOI: 10.1074/jbc.m116.756247] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/08/2016] [Indexed: 01/03/2023] Open
Abstract
TGF-β promotes excessive collagen deposition in fibrotic diseases such as idiopathic pulmonary fibrosis (IPF). The amino acid composition of collagen is unique due to its high (33%) glycine content. Here, we report that TGF-β induces expression of glycolytic genes and increases glycolytic flux. TGF-β also induces the expression of the enzymes of the de novo serine synthesis pathway (phosphoglycerate dehydrogenase (PHGDH), phosphoserine aminotransferase 1 (PSAT1), and phosphoserine phosphatase (PSPH)) and de novo glycine synthesis (serine hydroxymethyltransferase 2 (SHMT2)). Studies in fibroblasts with genetic attenuation of PHGDH or SHMT2 and pharmacologic inhibition of PHGDH showed that these enzymes are required for collagen synthesis. Furthermore, metabolic labeling experiments demonstrated carbon from glucose incorporated into collagen. Lungs from humans with IPF demonstrated increased expression of PHGDH and SHMT2. These results indicate that the de novo serine synthesis pathway is necessary for TGF-β-induced collagen production and suggest that this pathway may be a therapeutic target for treatment of fibrotic diseases including IPF.
Collapse
Affiliation(s)
- Recep Nigdelioglu
- From the Departments of Medicine, Section of Pulmonary and Critical Care Medicine, and
| | - Robert B Hamanaka
- From the Departments of Medicine, Section of Pulmonary and Critical Care Medicine, and
| | - Angelo Y Meliton
- From the Departments of Medicine, Section of Pulmonary and Critical Care Medicine, and
| | - Erin O'Leary
- From the Departments of Medicine, Section of Pulmonary and Critical Care Medicine, and
| | - Leah J Witt
- From the Departments of Medicine, Section of Pulmonary and Critical Care Medicine, and
| | - Takugo Cho
- From the Departments of Medicine, Section of Pulmonary and Critical Care Medicine, and
| | - Kaitlyn Sun
- From the Departments of Medicine, Section of Pulmonary and Critical Care Medicine, and
| | - Catherine Bonham
- From the Departments of Medicine, Section of Pulmonary and Critical Care Medicine, and
| | - David Wu
- From the Departments of Medicine, Section of Pulmonary and Critical Care Medicine, and
| | - Parker S Woods
- From the Departments of Medicine, Section of Pulmonary and Critical Care Medicine, and
| | | | - Don Wolfgeher
- University of Chicago Proteomics Core Facility, The University of Chicago, Chicago, Illinois 60637 and
| | - Nickolai O Dulin
- From the Departments of Medicine, Section of Pulmonary and Critical Care Medicine, and
| | - Navdeep S Chandel
- the Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois 60611
| | - Gökhan M Mutlu
- From the Departments of Medicine, Section of Pulmonary and Critical Care Medicine, and
| |
Collapse
|