1
|
Jiang P, Huang H, Liu Z, Xiang G, Wu X, Hao S, Li S. STAT6 deficiency mitigates the severity of pulmonary arterial hypertension caused by chronic intermittent hypoxia by suppressing Th2-inducing cytokines. Respir Res 2025; 26:13. [PMID: 39806384 PMCID: PMC11731530 DOI: 10.1186/s12931-024-03062-z] [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: 09/15/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is frequently associated with increased incidence and mortality of pulmonary hypertension (PH). The immune response contributes to pulmonary artery remodeling and OSA-related diseases. The immunologic factors linked to OSA-induced PH are not well understood. STAT6 is crucial in the signaling pathway that modulates immune response. However, the status of phosphorylated STAT6 (p-STAT6) in an OSA-induced PH mouse model remains largely unexplored. METHODS Chronic intermittent hypoxia (CIH) plays a crucial role in the progression of OSA. This study utilized a in vivo CIH model to examine the role of STAT6 in CIH-induced PH. RESULTS CIH mice exhibited pulmonary artery remodeling and pulmonary hypertension, indicated by increased right ventricular systolic pressure (RVSP), higher right ventricular to left ventricular plus septum (RV/LV + S) ratios, and significant morphological alterations compared to normoxic (Nor) mice. Increased p-STAT6 in the lungs and elevated p-STAT6 + IL-4 + producing T cells in CIH mice. STAT6 deficiency (STAT6-/-) improved PH and PA remodeling in CIH-induced PH mouse models.STAT6 deficiency impaired the T helper 2 (Th2) immune response, affecting IL-4 and IL-13 secretion. IL-4, rather than IL-13, activated STAT6 in human pulmonary artery smooth muscle cells (hPASMCs). STAT6 knockdown decreased the proliferation in IL-4 treated hPASMCs. CONCLUSION These findings exhibit the critical role of STAT6 in the pathogenesis of CIH induced PH by regulating Th2 immune response.STAT6 could be a significant therapeutic target for OSA-related PH.
Collapse
Affiliation(s)
- Pan Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- The Nutrition Department at Zhongshan Hospital, Fudan University, Shanghai, China
- The Nutrition Department, QingPu District Central Hospital, Shanghai, 200032, China
| | - Huai Huang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zilong Liu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guiling Xiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- The Critical Care Medicine Department at Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaodan Wu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Clinical Center for Sleep Breathing Disorder and Snoring, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Shengyu Hao
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- The Critical Care Medicine Department at Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Shanqun Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Clinical Center for Sleep Breathing Disorder and Snoring, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
Wang Y, Han D, Chai L, Qiu Y, Liu J, Li D, Zhang Q, Shen N, Chen Y, Chen H, Zhang J, Wang Q, Wang J, Li S, Xie X, Li M. MFN2-dependent mitochondrial dysfunction contributes to Relm-β-induced pulmonary arterial hypertension via USP18/Twist1/miR-214 pathway. Eur J Pharmacol 2024; 980:176828. [PMID: 39094924 DOI: 10.1016/j.ejphar.2024.176828] [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: 03/21/2024] [Revised: 07/03/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024]
Abstract
Induction of resistin-like molecule β (Relm-β) and mitofusin 2 (MFN2) mediated aberrant mitochondrial fission have been found to be involved in the pathogenesis of pulmonary arterial hypertension (PAH). However, the molecular mechanisms underlying Relm-β regulation of MFN2 therefore mitochondrial fission remain unclear. This study aims to address these issues. Primary cultured PASMCs and monocrotaline (MCT)-induced PAH rats were applied in this study. The results showed that Relm-β promoted cells proliferation in PASMCs, this was accompanied with the upregulation of USP18, Twist1 and miR-214, and downregulation of MFN2. We found that Relm-β increased USP18 expression which in turn raised Twist1 by suppressing its proteasome degradation. Elevation of Twist1 increased miR-214 expression and then reduced MFN2 expression and mitochondrial fragmentation leading to PASMCs proliferation. In vivo study, we confirmed that Relm-β was elevated in MCT-induced PAH rat model, and USP18/Twist1/miR-214/MFN2 axis was altered similar as in vitro. Targeting this cascade by Relm-β receptor inhibitor Calhex231, proteasome inhibitor MG-132, Twist1 inhibitor Harmine or miR-214 antagomiR prevented the development of pulmonary vascular remodeling and therefore PAH in MCT-treated rats. In conclusion, we demonstrate that Relm-β promotes PASMCs proliferation and vascular remodeling by activating USP18/Twist1/miR-214 dependent MFN2 reduction and mitochondrial fission, suggesting that this signaling pathway might be a promising target for management of PAH.
Collapse
Affiliation(s)
- Yan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Dong Han
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, PR China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Yuanjie Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Jin Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Danyang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Nirui Shen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Yuqian Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Huan Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Jia Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Xinming Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
| |
Collapse
|
3
|
Liu D, Zeng F, Chen Z, Qin Z, Liu Z. Regulation of cardiac fibrosis in mice with TAC/DOCA-induced HFpEF by resistin-like molecule gamma and adenylate cyclase 1. FEBS Open Bio 2024; 14:1101-1115. [PMID: 38710658 PMCID: PMC11216931 DOI: 10.1002/2211-5463.13813] [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: 06/06/2023] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/08/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is one of the major subtypes of heart failure (HF) and no effective treatments for this common disease exist to date. Cardiac fibrosis is central to the pathology of HF and a potential avenue for the treatment of HFpEF. To explore key fibrosis-related genes and pathways in the pathophysiological process of HFpEF, a mouse model of HFpEF was constructed. The relevant gene expression profiles were downloaded from the Gene Expression Omnibus database, and single-sample Gene Set Enrichment Analysis (ssGSEA) was performed targeting fibrosis-related pathways to explore differentially expressed genes (DEGs) in healthy control and HFpEF heart tissues with cross-tabulation analysis of fibrosis-related genes. Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed on the identified fibrosis-related genes. The two most significant DEGs were selected, and further validation was conducted in HFpEF mice. The results indicated that myocardial fibrosis was significantly upregulated in HFpEF mice compared to healthy controls, while the ssGSEA results revealed significant differences in the enrichment of nine fibrosis-related pathways in HFpEF myocardial tissue, with 112 out of 798 DEGs being related to fibrosis. The in vivo results demonstrated that expression levels of resistin-like molecule gamma (Relmg) and adenylate cyclase 1 (Adcy1) in the heart tissues of HFpEF mice were significantly higher and lower, respectively, compared to healthy controls. Taken together, these results suggest that Relmg and Acdy1 as well as the fibrosis process may be potential targets for HFpEF treatment.
Collapse
Affiliation(s)
- Dawei Liu
- The First Affiliated Hospital of Chongqing Medical UniversityChina
- Department of Cardiology, Bishan Hospital of ChongqingBishan Hospital of Chongqing Medical UniversityChina
| | - Fanling Zeng
- Health Management CenterThe First Affiliated Hospital of Chongqing Medical UniversityChina
| | - Zhiyu Chen
- Orthopedic Laboratory of Chongqing Medical UniversityChina
| | - Zheng Qin
- Department of Vascular SurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChina
| | - Zhiqiang Liu
- The First Affiliated Hospital of Chongqing Medical UniversityChina
| |
Collapse
|
4
|
Zuo Y, Li B, Gao M, Xiong R, He R, Li N, Geng Q. Novel insights and new therapeutic potentials for macrophages in pulmonary hypertension. Respir Res 2024; 25:147. [PMID: 38555425 PMCID: PMC10981837 DOI: 10.1186/s12931-024-02772-8] [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: 07/23/2023] [Accepted: 03/13/2024] [Indexed: 04/02/2024] Open
Abstract
Inflammation and immune processes underlie pulmonary hypertension progression. Two main different activated phenotypes of macrophages, classically activated M1 macrophages and alternatively activated M2 macrophages, are both involved in inflammatory processes related to pulmonary hypertension. Recent advances suggest that macrophages coordinate interactions among different proinflammatory and anti-inflammatory mediators, and other cellular components such as smooth muscle cells and fibroblasts. In this review, we summarize the current literature on the role of macrophages in the pathogenesis of pulmonary hypertension, including the origin of pulmonary macrophages and their response to triggers of pulmonary hypertension. We then discuss the interactions among macrophages, cytokines, and vascular adventitial fibroblasts in pulmonary hypertension, as well as the potential therapeutic benefits of macrophages in this disease. Identifying the critical role of macrophages in pulmonary hypertension will contribute to a comprehensive understanding of this pathophysiological abnormality, and may provide new perspectives for pulmonary hypertension management.
Collapse
Affiliation(s)
- Yifan Zuo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Boyang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Minglang Gao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Rui Xiong
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
5
|
Hanyu F, Zheng H, Jiaqi W, Tairan D, Yiyuanzi Z, Qiwen Y, Ying L, Hongchun Z, Lu L. Protective effects and mechanism of curcumin in animal models of pulmonary fibrosis: a preclinical systematic review and meta-analysis. Front Pharmacol 2023; 14:1258885. [PMID: 37900163 PMCID: PMC10613035 DOI: 10.3389/fphar.2023.1258885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/28/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction: At present, there is a lack of effective treatment for pulmonary fibrosis (PF), and a number of studies have confirmed that curcumin (CUR) has a good effect on PF. Research Qusetion: Is CUR effective in preclinical trials for PF and what is its mechanism of action? Methods: Animal reports of PF treated with CUR were searched from Pubmed, Embase, Web of Science and Cochrane Library from 1 January 2000 to 19 April 2023 to compare CUR treatment of PF with a no-intervention model group. A previous registration (nsply registration number: INPLASY202360084) of this review protocol was undertaken. Results: The meta-analysis included 27 publications and 29 studies involving 396 animals. CUR significantly improved the degree of fibrosis, levels of inflammation, and oxidative imbalances in lung tissue in animal models of PF. In terms fibrosis, such as HYP content (SMD = -4.96; 95% CI = -6.05 to -3.87; p = 0.000).In terms of inflammatory indicators, such as MPO activity (SMD = -2.12; 95% CI = -4.93 to 0.69; p = 0.000). In terms of oxidation index, such as MDA (SMD = -5.63; 95% CI = -9.66 to -1.6; p = 0.000). Conclusion: CUR significantly improved the degree of fibrosis, levels of inflammation, and oxidative imbalances in lung tissue in animal models of PF. Due to the quantitative and qualitative limitations of current research, more high-quality studies are needed to verify the above conclusion.
Collapse
Affiliation(s)
- Fang Hanyu
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Hong Zheng
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Wang Jiaqi
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Dong Tairan
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Zhao Yiyuanzi
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Yang Qiwen
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Liu Ying
- The Second Health and Medical Department, China-Japan Friendship Hospital, Beijing, China
| | - Zhang Hongchun
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
- Department of Traditional Chinese Medicine for Pulmonary Diseases, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Liu Lu
- Department of Traditional Chinese Medicine for Pulmonary Diseases, Jining Hospital of Xiyuan Hospital of China Academy of Chinese Medical Science, Jining, Shandong, China
| |
Collapse
|
6
|
Che L, Xie Z, Chen G, Zhang W, Xia T, Lin J, Luo W, Chen L, Yin W, Cai X, Liu S. The Mechanisms of Resistin-Like Molecule-β-Mediated Airway Inflammation in Chronic Obstructive Pulmonary Disease via Autophagy. J Inflamm Res 2023; 16:3853-3870. [PMID: 37671130 PMCID: PMC10476668 DOI: 10.2147/jir.s403517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 08/01/2023] [Indexed: 09/07/2023] Open
Abstract
Background The role of irreversible airway inflammatory damage in chronic obstructive pulmonary disease (COPD) progression is evident. Autophagy is an essential process in the cellular material metabolic cycle, and a family of resistant vegetative molecules may be involved in the COPD autophagic process. In this study, we investigated the mechanism of resistin-like molecule β (RELMβ) in COPD smoking-induced autophagy. Methods Firstly, the expression differences of RELMβ and autophagy markers between COPD and control groups were analyzed in the Gene Expression Omnibus (GEO) datasets and clinical specimens. Secondly, in vitro and in vivo experiments were conducted using immunoblotting, immunofluorescence, immunohistochemistry, and other methods to investigate the mechanism by which RELMβ promotes airway inflammation through autophagy in a cigarette smoke extract-induced 16HBE cell inflammation model and a cigarette smoke-induced COPD-like mouse model. In addition, immunoprecipitation was used to analyze the binding of RELMβ to the membrane protein TLR4. Results The expression of RELMβ and autophagy genes p62 and LC3B in lung tissue of COPD patients was significantly increased. RELMβ can mediate the activation of autophagy in 16HBE cells, and through autophagy, it increases the expression of inflammatory cytokines in a cigarette smoke extract-induced 16HBE cell inflammation model. RELMβ promotes cigarette smoke-induced COPD-like mouse airway inflammation through autophagy, and RELMβ can mediate signal transduction through the cell membrane receptor TLR4. Conclusion The RELMβ binds to TLR4 to encourage signal transduction and that RELMβ can promote inflammation in smoky COPD lungs through autophagy.
Collapse
Affiliation(s)
- Li Che
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Zhefan Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
- Affiliated Dongguan People’s Hospital, Southern Medical University, Donguan, People’s Republic of China
| | - Guangshu Chen
- Department of Endocrinology, Guangzhou Red Cross Hospital, The Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Wei Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Tingting Xia
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
- Affiliated Dongguan People’s Hospital, Southern Medical University, Donguan, People’s Republic of China
| | - Jiaxin Lin
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Wenzhi Luo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Li Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Wenguang Yin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Xingdong Cai
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Shengming Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| |
Collapse
|
7
|
Lin Q, Kumar S, Kariyawasam U, Yang X, Yang W, Skinner JT, Gao WD, Johns RA. Human Resistin Induces Cardiac Dysfunction in Pulmonary Hypertension. J Am Heart Assoc 2023; 12:e027621. [PMID: 36927008 PMCID: PMC10111547 DOI: 10.1161/jaha.122.027621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 03/18/2023]
Abstract
Background Cardiac failure is the primary cause of death in most patients with pulmonary arterial hypertension (PH). As pleiotropic cytokines, human resistin (Hresistin) and its rodent homolog, resistin-like molecule α, are mechanistically critical to pulmonary vascular remodeling in PH. However, it is still unclear whether activation of these resistin-like molecules can directly cause PH-associated cardiac dysfunction and remodeling. Methods and Results In this study, we detected Hresistin protein in right ventricular (RV) tissue of patients with PH and elevated resistin-like molecule expression in RV tissues of rodents with RV hypertrophy and failure. In a humanized mouse model, cardiac-specific Hresistin overexpression was sufficient to cause cardiac dysfunction and remodeling. Dilated hearts exhibited reduced force development and decreased intracellular Ca2+ transients. In the RV tissues overexpressing Hresistin, the impaired contractility was associated with the suppression of protein kinase A and AMP-activated protein kinase. Mechanistically, Hresistin activation triggered the inflammation mediated by signaling of the key damage-associated molecular pattern molecule high-mobility group box 1, and subsequently induced pro-proliferative Ki67 in RV tissues of the transgenic mice. Intriguingly, an anti-Hresistin human antibody that we generated protected the myocardium from hypertrophy and failure in the rodent PH models. Conclusions Our data indicate that Hresistin is expressed in heart tissues and plays a role in the development of RV dysfunction and maladaptive remodeling through its immunoregulatory activities. Targeting this signaling to modulate cardiac inflammation may offer a promising strategy to treat PH-associated RV hypertrophy and failure in humans.
Collapse
Affiliation(s)
- Qing Lin
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Santosh Kumar
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Udeshika Kariyawasam
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Xiaomei Yang
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
- Department of AnesthesiologyQilu Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
| | - Wei Yang
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
- Department of Cardiovascular MedicineXiangya Hospital, Central South UniversityChangshaChina
| | - John T. Skinner
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Roger A. Johns
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| |
Collapse
|
8
|
Nappi F, Avtaar Singh SS. Distinctive Signs of Disease as Deterrents for the Endothelial Function: A Systematic Review. Metabolites 2023; 13:metabo13030430. [PMID: 36984870 PMCID: PMC10057506 DOI: 10.3390/metabo13030430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Endothelial integrity plays a major role in homeostasis and is responsive to the numerous endogenous factors released. While its functional role in vascular tone is well described, its role in the pathophysiology of cardiovascular disease is of interest as a potential therapeutic target. We performed a systematic review to provide an overview of new therapeutic and diagnostic targets for the treatment of coronary artery disease related to endothelial dysfunction. Databases of PubMed, Ovid’s version of MEDLINE, and EMBASE were interrogated with appropriate search terms. Inclusion criteria have been met by 28 studies that were included in the final systematic review. We identified inflammation, pulmonary hypertension, diabetes mellitus and Fabry disease as pathophysiological mechanisms and explored the therapeutic options related to these conditions including medications such as Canakinumab. Endothelial dysfunction has a key role in several different pathophysiological processes which can be targeted for therapeutic options. Ongoing research should be targeted at making the transition to clinical practice. Further research is also needed on understanding the amelioration of endothelial dysfunction with the use of cardiovascular medications.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
- Correspondence: ; Tel.: +33-149334104; Fax: +33-149334119
| | | |
Collapse
|
9
|
Shi Y, Zhu N, Qiu Y, Tan J, Wang F, Qin L, Dai A. Resistin-like molecules: a marker, mediator and therapeutic target for multiple diseases. Cell Commun Signal 2023; 21:18. [PMID: 36691020 PMCID: PMC9869618 DOI: 10.1186/s12964-022-01032-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/27/2022] [Indexed: 01/25/2023] Open
Abstract
Resistin-like molecules (RELMs) are highly cysteine-rich proteins, including RELMα, RELMβ, Resistin, and RELMγ. However, RELMs exhibit significant differences in structure, distribution, and function. The expression of RELMs is regulated by various signaling molecules, such as IL-4, IL-13, and their receptors. In addition, RELMs can mediate numerous signaling pathways, including HMGB1/RAGE, IL-4/IL-4Rα, PI3K/Akt/mTOR signaling pathways, and so on. RELMs proteins are involved in wide range of physiological and pathological processes, including inflammatory response, cell proliferation, glucose metabolism, barrier defense, etc., and participate in the progression of numerous diseases such as lung diseases, intestinal diseases, cardiovascular diseases, and cancers. Meanwhile, RELMs can serve as biomarkers, risk predictors, and therapeutic targets for these diseases. An in-depth understanding of the role of RELMs may provide novel targets or strategies for the treatment and prevention of related diseases. Video abstract.
Collapse
Affiliation(s)
- Yaning Shi
- Laboratory of Stem Cell Regulation with Chinese Medicine and its Application, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
- Science and Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410021, Hunan, China
| | - Yun Qiu
- Laboratory of Stem Cell Regulation with Chinese Medicine and its Application, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Junlan Tan
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, 410208, Hunan, China
| | - Feiying Wang
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, 410208, Hunan, China
| | - Li Qin
- Laboratory of Stem Cell Regulation with Chinese Medicine and its Application, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, 410208, Hunan, China.
| | - Aiguo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, 410208, Hunan, China.
- Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
- Department of Respiratory Medicine, First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410021, Hunan, China.
| |
Collapse
|
10
|
Shin H, Morty RE, Sucre JM, Negretti NM, Markmann M, Hossain H, Krauss-Etschmann S, Dehmel S, Hilgendorff A. Reference genes for the developing mouse lung under consideration of biological, technical and experimental confounders. Sci Rep 2022; 12:17679. [PMID: 36271035 PMCID: PMC9587035 DOI: 10.1038/s41598-022-19071-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/24/2022] [Indexed: 01/18/2023] Open
Abstract
For gene expression analysis, the raw data obtained from RT-qPCR are preferably normalized to reference genes, which should be constantly expressed regardless of experimental conditions. Selection of reference genes is particularly challenging for the developing lung because of the complex transcriptional and epigenetic regulation of genes during organ maturation and injury repair. To date, there are only limited experimental data addressing reliable reference genes for this biological circumstance. In this study, we evaluated reference genes for the lung in neonatal C57BL/6 mice under consideration of biological, technical and experimental conditions. For that, we thoroughly selected candidates from commonly used reference genes side-by-side with novel ones by analyzing publicly available microarray datasets. We performed RT-qPCR of the selected candidate genes and analyzed their expression variability using GeNorm and Normfinder. Cell-specific expression of the candidate genes was analyzed using our own single-cell RNA-sequencing data from the developing mouse lung. Depending on the investigated conditions, i.e., developmental stages, sex, RNA quality, experimental condition (hyperoxia) and cell types, distinct candidate genes demonstrated stable expression confirming their eligibility as reliable reference genes. Our results provide valuable information for the selection of proper reference genes in studies investigating the neonatal mouse lung.
Collapse
Affiliation(s)
- H. Shin
- grid.4567.00000 0004 0483 2525Institute for Lung Biology and Disease and Comprehensive Pneumology Center, Helmholtz Zentrum München, Member of German Center for Lung Research (DZL), Munich, Germany
| | - R. E. Morty
- grid.5253.10000 0001 0328 4908Department of Translational Pulmonology, University Hospital Heidelberg, Heidelberg, Germany ,Translational Lung Research Center, member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - J. M. Sucre
- grid.412807.80000 0004 1936 9916Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN USA
| | - N. M. Negretti
- grid.412807.80000 0004 1936 9916Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN USA
| | - M. Markmann
- grid.8664.c0000 0001 2165 8627Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Justus-Liebig-University, Giessen, Germany
| | - H. Hossain
- grid.440273.6Institute of Laboratory Medicine and Microbiology, Klinikum St. Marien Amberg and Kliniken Nordoberpfalz AG, Weiden, Germany
| | - S. Krauss-Etschmann
- grid.4567.00000 0004 0483 2525Institute for Lung Biology and Disease and Comprehensive Pneumology Center, Helmholtz Zentrum München, Member of German Center for Lung Research (DZL), Munich, Germany ,grid.452624.3Present Address: Priority Area Chronic Lung Diseases, Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, German Center for Lung Research (DZL) and the Airway Research Center North (ARCN), Borstel, Germany ,grid.9764.c0000 0001 2153 9986Present Address: Institute for Experimental Medicine, Christian Albrechts University, German Center for Lung Research (DZL) and the Airway Research Center North (ARCN), Kiel, Germany
| | - S. Dehmel
- grid.4567.00000 0004 0483 2525Institute for Lung Biology and Disease and Comprehensive Pneumology Center, Helmholtz Zentrum München, Member of German Center for Lung Research (DZL), Munich, Germany ,grid.4567.00000 0004 0483 2525Present Address: Strategy, Programs, Resources (SPR), Helmholtz Zentrum München, Munich, Germany
| | - A. Hilgendorff
- grid.4567.00000 0004 0483 2525Institute for Lung Biology and Disease and Comprehensive Pneumology Center, Helmholtz Zentrum München, Member of German Center for Lung Research (DZL), Munich, Germany ,grid.5252.00000 0004 1936 973XCenter for Comprehensive Developmental Care (CDeCLMU), University Hospital, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
11
|
Tatu AL, Nadasdy T, Arbune A, Chioncel V, Bobeica C, Niculet E, Iancu AV, Dumitru C, Popa VT, Kluger N, Clatici VG, Vasile CI, Onisor C, Nechifor A. Interrelationship and Sequencing of Interleukins4, 13, 31, and 33 - An Integrated Systematic Review: Dermatological and Multidisciplinary Perspectives. J Inflamm Res 2022; 15:5163-5184. [PMID: 36110506 PMCID: PMC9468867 DOI: 10.2147/jir.s374060] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/13/2022] [Indexed: 11/23/2022] Open
Abstract
The interrelations and sequencing of interleukins are complex (inter)actions where each interleukin can stimulate the secretion of its preceding interleukin. In this paper, we attempt to summarize the currently known roles of IL-4, IL-13, IL-31, and IL-33 from a multi-disciplinary perspective. In order to conduct a comprehensive review of the current literature, a search was conducted using PubMed, Google Scholar, Medscape, UpToDate, and Key Elsevier for keywords. The results were compiled from case reports, case series, letters, and literature review papers, and analyzed by a panel of multi-disciplinary specialist physicians for relevance. Based on 173 results, we compiled the following review of interleukin signaling and its clinical significance across a multitude of medical specialties. Interleukins are at the bed rock of a multitude of pathologies across different organ systems and understanding their role will likely lead to novel treatments and better outcomes for our patients. New interleukins are being described, and the role of this inflammatory cascade is still coming to light. We hope this multi-discipline review on the role interleukins play in current pathology assists in this scope.
Collapse
Affiliation(s)
- Alin Laurentiu Tatu
- Dermatology Department, "Sf. Cuvioasa Parascheva" Clinical Hospital of Infectious Diseases, Galati, Romania.,Clinical Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Galati, Romania.,Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR) [Centrul Integrat Multi disciplinar de Cercetare de Interfata Dermatologica (CIM-CID)], Galați, Romania
| | - Thomas Nadasdy
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR) [Centrul Integrat Multi disciplinar de Cercetare de Interfata Dermatologica (CIM-CID)], Galați, Romania.,Dermatology Department, Municipal Emergency Hospital, Timişoara, Romania
| | - Anca Arbune
- Neurology Department, Fundeni Clinical Institute, Bucharest, Romania
| | - Valentin Chioncel
- Neurology Department, "Bagdasar-Arseni" Emergency Clinical Hospital, Bucharest, Romania
| | - Carmen Bobeica
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galați, Romania
| | - Elena Niculet
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR) [Centrul Integrat Multi disciplinar de Cercetare de Interfata Dermatologica (CIM-CID)], Galați, Romania
| | - Alina Viorica Iancu
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galați, Romania
| | - Caterina Dumitru
- Pharmaceutical Sciences Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Galati, Romania
| | - Valentin Tudor Popa
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR) [Centrul Integrat Multi disciplinar de Cercetare de Interfata Dermatologica (CIM-CID)], Galați, Romania.,Dermatology Department, Center for the Morphologic Study of the Skin MORPHODERM, "Victor Babeș" University of Medicine and Pharmacy, Timișoara, Romania
| | - Nicolas Kluger
- Department of Dermatology, Allergology and Venereology, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland.,Apolo Medical Center, Bucharest, Romania
| | | | - Claudiu Ionut Vasile
- Clinical Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Galati, Romania
| | - Cristian Onisor
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galați, Romania
| | - Alexandru Nechifor
- Clinical Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Galati, Romania
| |
Collapse
|
12
|
Li Y, Zhang Q, Li L, Hao D, Cheng P, Li K, Li X, Wang J, Wang Q, Du Z, Ji H, Chen H. LKB1 deficiency upregulates RELM-α to drive airway goblet cell metaplasia. Cell Mol Life Sci 2021; 79:42. [PMID: 34921639 PMCID: PMC8738459 DOI: 10.1007/s00018-021-04044-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 02/08/2023]
Abstract
Targeting airway goblet cell metaplasia is a novel strategy that can potentially reduce the chronic obstructive pulmonary disease (COPD) symptoms. Tumor suppressor liver kinase B1 (LKB1) is an important regulator of the proliferation and differentiation of stem/progenitor cells. In this study, we report that LKB1 expression was downregulated in the lungs of patients with COPD and in those of cigarette smoke-exposed mice. Nkx2.1Cre; Lkb1f/f mice with conditional loss of Lkb1 in mouse lung epithelium displayed airway mucus hypersecretion and pulmonary macrophage infiltration. Single-cell transcriptomic analysis of the lung tissues from Nkx2.1Cre; Lkb1f/f mice further revealed that airway goblet cell differentiation was altered in the absence of LKB1. An organoid culture study demonstrated that Lkb1 deficiency in mouse airway (club) progenitor cells promoted the expression of FIZZ1/RELM-α, which drove airway goblet cell differentiation and pulmonary macrophage recruitment. Additionally, monocyte-derived macrophages in the lungs of Nkx2.1Cre; Lkb1f/f mice exhibited an alternatively activated M2 phenotype, while expressing RELM-α, which subsequently aggravated airway goblet cell metaplasia. Our findings suggest that the LKB1-mediated crosstalk between airway progenitor cells and macrophages regulates airway goblet cell metaplasia. Moreover, our data suggest that LKB1 agonists might serve as a potential therapeutic option to treat respiratory disorders associated with goblet cell metaplasia.
Collapse
Affiliation(s)
- Yu Li
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| | - Qiuyang Zhang
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| | - Li Li
- Department of Respiratory Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
| | - De Hao
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
| | - Peiyong Cheng
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
| | - Kuan Li
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| | - Xue Li
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| | - Jianhai Wang
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| | - Qi Wang
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
| | - Zhongchao Du
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
| | - Hongbin Ji
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Huaiyong Chen
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, 300350, China.
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China.
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China.
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China.
| |
Collapse
|
13
|
Che L, Yu C, Chen G, Lin J, Xie Z, Xia T, Luo W, Cai X, Liu S. The Inflammatory Response Induced by RELMβ Upregulates IL-8 and IL-1β Expression in Bronchial Epithelial Cells in COPD. Int J Chron Obstruct Pulmon Dis 2021; 16:2503-2513. [PMID: 34511895 PMCID: PMC8421257 DOI: 10.2147/copd.s321877] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/23/2021] [Indexed: 01/04/2023] Open
Abstract
Purpose Chronic obstructive pulmonary disease (COPD) is associated with a complex inflammatory regulatory network. Resistin-like molecule β (RELMβ) is highly expressed in the lungs of COPD patients. We aimed to investigate the proinflammatory effect of RELMβ on airway epithelial cells in COPD. Methods First, a GEO dataset was used to analyze the expression of the RELMβ gene in the COPD and control groups as well as the protein levels of RELMβ in the sera of outpatients with COPD and normal control subjects in our hospital. We also stimulated 16HBE bronchial epithelial cells with recombinant RELMβ protein and analyzed the expression of IL-8 and IL-1β. We upregulated and downregulated the gene expression of RELMβ in 16HBE cells and analyzed the expression of the inflammatory cytokines IL-8 and IL-1β. In addition, we also examined the mechanism by which the p38 MAPK signaling pathway contributed to the regulation of IL-8 and IL-1β expression by RELMβ. Results RELMβ expression was increased in COPD tissues in different data sets and in the serum of COPD patients in our hospital. IL-8 and IL-1β expression was also increased in COPD tissues with high RELMβ gene expression in different data sets. The RELMβ gene was mainly related to inflammatory factors and inflammatory signaling pathways in the PPI regulatory network. Experiments at the cellular level showed that RELMβ promoted the expression of the inflammatory cytokines IL-8 and IL-1β, and this regulation was mediated by the p38 MAPK signaling pathway. Conclusion RELMβ can promote the expression of the inflammatory cytokines IL-8 and IL-1β in bronchial epithelial cells of patients with COPD and exert inflammatory effects. RELMβ may be a potential target for the treatment of COPD.
Collapse
Affiliation(s)
- Li Che
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, People's Republic of China
| | - Chao Yu
- Department of Pulmonary and Critical Care Medicine, Lu'an People's Hospital of Anhui Province, Lu'an, 237016, People's Republic of China
| | - Guangshu Chen
- Department of Endocrinology, Guangzhou Red Cross Hospital, The Affiliated Hospital of Jinan University, Guangzhou, 510220, People's Republic of China
| | - Jiaxin Lin
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, People's Republic of China
| | - Zhefan Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, People's Republic of China
| | - Tingting Xia
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, People's Republic of China
| | - Wenzhi Luo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, People's Republic of China
| | - Xingdong Cai
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, People's Republic of China
| | - Shengming Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, People's Republic of China
| |
Collapse
|
14
|
Role of the Immune System Elements in Pulmonary Arterial Hypertension. J Clin Med 2021; 10:jcm10163757. [PMID: 34442052 PMCID: PMC8397145 DOI: 10.3390/jcm10163757] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/11/2021] [Accepted: 08/20/2021] [Indexed: 02/08/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a relatively rare disease, but, today, its incidence tends to increase. The severe course of the disease and poor patient survival rate make PAH a major diagnostic and therapeutic challenge. For this reason, a thorough understanding of the pathogenesis of the disease is essential to facilitate the development of more effective therapeutic targets. Research shows that the development of PAH is characterized by a number of abnormalities within the immune system that greatly affect the progression of the disease. In this review, we present key data on the regulated function of immune cells, released cytokines and immunoregulatory molecules in the development of PAH, to help improve diagnosis and targeted immunotherapy.
Collapse
|
15
|
Nakahara M, Ito H, Skinner JT, Lin Q, Tamosiuniene R, Nicolls MR, Keegan AD, Johns RA, Yamaji-Kegan K. The inflammatory role of dysregulated IRS2 in pulmonary vascular remodeling under hypoxic conditions. Am J Physiol Lung Cell Mol Physiol 2021; 321:L416-L428. [PMID: 34189964 PMCID: PMC8410109 DOI: 10.1152/ajplung.00068.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/24/2022] Open
Abstract
Pulmonary hypertension (PH) is a devastating disease characterized by progressive elevation of pulmonary vascular resistance, right ventricular failure, and ultimately death. We have shown previously that insulin receptor substrate 2 (IRS2), a molecule highly critical to insulin resistance and metabolism, has an anti-inflammatory role in Th2-skewed lung inflammation and pulmonary vascular remodeling. Here, we investigated the hypothesis that IRS2 has an immunomodulatory role in human and experimental PH. Expression analysis showed that IRS2 was significantly decreased in the pulmonary vasculature of patients with pulmonary arterial hypertension and in rat models of PH. In mice, genetic ablation of IRS2 enhanced the hypoxia-induced signaling pathway of Akt and Forkhead box O1 (FOXO1) in the lung tissue and increased pulmonary vascular muscularization, proliferation, and perivascular macrophage recruitment. Furthermore, mice with homozygous IRS2 gene deletion showed a significant gene dosage-dependent increase in pulmonary vascular remodeling and right ventricular hypertrophy in response to hypoxia. Functional studies with bone marrow-derived macrophages isolated from homozygous IRS2 gene-deleted mice showed that hypoxia exposure led to enhancement of the Akt and ERK signaling pathway followed by increases in the pro-PH macrophage activation markers, vascular endothelial growth factor-A and arginase 1. Our data suggest that IRS2 contributes to anti-inflammatory effects by regulating macrophage activation and recruitment, which may limit the vascular inflammation, remodeling, and right ventricular hypertrophy that are seen in PH pathology. Restoring the IRS2 pathway may be an effective therapeutic approach for the treatment of PH and right heart failure.
Collapse
Affiliation(s)
- Mayumi Nakahara
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Homare Ito
- Department of Anesthesiology, University of Maryland Baltimore, Baltimore, Maryland
| | - John T Skinner
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Qing Lin
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Rasa Tamosiuniene
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California
| | - Mark R Nicolls
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California
| | - Achsah D Keegan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Roger A Johns
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Kazuyo Yamaji-Kegan
- Department of Anesthesiology, University of Maryland Baltimore, Baltimore, Maryland
| |
Collapse
|
16
|
Lv M, Liu W. Hypoxia-Induced Mitogenic Factor: A Multifunctional Protein Involved in Health and Disease. Front Cell Dev Biol 2021; 9:691774. [PMID: 34336840 PMCID: PMC8319639 DOI: 10.3389/fcell.2021.691774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/23/2021] [Indexed: 11/13/2022] Open
Abstract
Hypoxia-induced mitogenic factor (HIMF), also known as resistin-like molecule α (RELMα) or found in inflammatory zone 1 (FIZZ1) is a member of the RELM protein family expressed in mice. It is involved in a plethora of physiological processes, including mitogenesis, angiogenesis, inflammation, and vasoconstriction. HIMF expression can be stimulated under pathological conditions and this plays a critical role in pulmonary, cardiovascular and metabolic disorders. The present review summarizes the molecular characteristics, and the physiological and pathological roles of HIMF in normal and diseased conditions. The potential clinical significance of these findings for human is also discussed.
Collapse
Affiliation(s)
- Moyang Lv
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Wenjuan Liu
- Department of Pathophysiology, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
17
|
Tao B, Kumar S, Gomez-Arroyo J, Fan C, Zhang A, Skinner J, Hunter E, Yamaji-Kegan K, Samad I, Hillel AT, Lin Q, Zhai W, Gao WD, Johns RA. Resistin-Like Molecule α Dysregulates Cardiac Bioenergetics in Neonatal Rat Cardiomyocytes. Front Cardiovasc Med 2021; 8:574708. [PMID: 33981729 PMCID: PMC8107692 DOI: 10.3389/fcvm.2021.574708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 03/30/2021] [Indexed: 11/13/2022] Open
Abstract
Heart (right) failure is the most frequent cause of death in patients with pulmonary arterial hypertension. Although historically, increased right ventricular afterload has been considered the main contributor to right heart failure in such patients, recent evidence has suggested a potential role of load-independent factors. Here, we tested the hypothesis that resistin-like molecule α (RELMα), which has been implicated in the pathogenesis of vascular remodeling in pulmonary artery hypertension, also contributes to cardiac metabolic remodeling, leading to heart failure. Recombinant RELMα (rRELMα) was generated via a Tet-On expression system in the T-REx 293 cell line. Cultured neonatal rat cardiomyocytes were treated with purified rRELMα for 24 h at a dose of 50 nM. Treated cardiomyocytes exhibited decreased mRNA and protein expression of peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α) and transcription factors PPARα and ERRα, which regulate mitochondrial fatty acid metabolism, whereas genes that encode for glycolysis-related proteins were significantly upregulated. Cardiomyocytes treated with rRELMα also exhibited a decreased basal respiration, maximal respiration, spare respiratory capacity, ATP-linked OCR, and increased glycolysis, as assessed with a microplate-based cellular respirometry apparatus. Transmission electron microscopy revealed abnormal mitochondrial ultrastructure in cardiomyocytes treated with rRELMα. Our data indicate that RELMα affects cardiac energy metabolism and mitochondrial structure, biogenesis, and function by downregulating the expression of the PGC-1α/PPARα/ERRα axis.
Collapse
Affiliation(s)
- Bingdong Tao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Santosh Kumar
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Jose Gomez-Arroyo
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Chunling Fan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Ailan Zhang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - John Skinner
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Elizabeth Hunter
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Kazuyo Yamaji-Kegan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
- Department of Anesthesiology, Maryland University, School of Medicine, Baltimore, MD, United States
| | - Idris Samad
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Alexander T. Hillel
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Qing Lin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Wenqian Zhai
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
- Department of Anesthesiology, Tianjin Chest Hospital, Tianjin, China
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Roger A. Johns
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| |
Collapse
|
18
|
Pai S, Njoku DB. The Role of Hypoxia-Induced Mitogenic Factor in Organ-Specific Inflammation in the Lung and Liver: Key Concepts and Gaps in Knowledge Regarding Molecular Mechanisms of Acute or Immune-Mediated Liver Injury. Int J Mol Sci 2021; 22:ijms22052717. [PMID: 33800244 PMCID: PMC7962531 DOI: 10.3390/ijms22052717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 01/15/2023] Open
Abstract
Hypoxia-induced mitogenic factor (HIMF), which is also known as resistin-like molecule α (RELM-α), found in inflammatory zone 1 (FIZZ1), or resistin-like alpha (retlna), is a cysteine-rich secretory protein and cytokine. HIMF has been investigated in the lung as a mediator of pulmonary fibrosis, inflammation and as a marker for alternatively activated macrophages. Although these macrophages have been found to have a role in acute liver injury and acetaminophen toxicity, few studies have investigated the role of HIMF in acute or immune-mediated liver injury. The aim of this focused review is to analyze the literature and examine the effects of HIMF and its human homolog in organ-specific inflammation in the lung and liver. We followed the guidelines set by PRISMA in constructing this review. The relevant checklist items from PRISMA were included. Items related to meta-analysis were excluded because there were no randomized controlled clinical trials. We found that HIMF was increased in most models of acute liver injury and reduced damage from acetaminophen-induced liver injury. We also found strong evidence for HIMF as a marker for alternatively activated macrophages. Our overall risk of bias assessment of all studies included revealed that 80% of manuscripts demonstrated some concerns in the randomization process. We also demonstrated some concerns (54.1%) and high risk (45.9%) of bias in the selection of the reported results. The need for randomization and reduction of bias in the reported results was similarly detected in the studies that focused on HIMF and the liver. In conclusion, we propose that HIMF could be utilized as a marker for M2 macrophages in immune-mediated liver injury. However, we also detected the need for randomized clinical trials and additional experimental and human prospective studies in order to fully comprehend the role of HIMF in acute or immune-mediated liver injury.
Collapse
Affiliation(s)
- Sananda Pai
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21287, USA;
| | - Dolores B. Njoku
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21287, USA;
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD 21287, USA
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21287, USA
- Correspondence:
| |
Collapse
|
19
|
Tian H, Liu L, Wu Y, Wang R, Jiang Y, Hu R, Zhu L, Li L, Fang Y, Yang C, Ji L, Liu G, Dai A. Resistin-like molecule β acts as a mitogenic factor in hypoxic pulmonary hypertension via the Ca 2+-dependent PI3K/Akt/mTOR and PKC/MAPK signaling pathways. Respir Res 2021; 22:8. [PMID: 33407472 PMCID: PMC7789700 DOI: 10.1186/s12931-020-01598-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 12/09/2020] [Indexed: 12/28/2022] Open
Abstract
Background Pulmonary arterial smooth muscle cell (PASMC) proliferation plays a crucial role in hypoxia-induced pulmonary hypertension (HPH). Previous studies have found that resistin-like molecule β (RELM-β) is upregulated de novo in response to hypoxia in cultured human PASMCs (hPASMCs). RELM-β has been reported to promote hPASMC proliferation and is involved in pulmonary vascular remodeling in patients with PAH. However, the expression pattern, effects, and mechanisms of action of RELM-β in HPH remain unclear. Methods We assessed the expression pattern, mitogenetic effect, and mechanism of action of RELM-β in a rat HPH model and in hPASMCs. Results Overexpression of RELM-β caused hemodynamic changes in a rat model of HPH similar to those induced by chronic hypoxia, including increased mean right ventricular systolic pressure (mRVSP), right ventricular hypertrophy index (RVHI) and thickening of small pulmonary arterioles. Knockdown of RELM-β partially blocked the increases in mRVSP, RVHI, and vascular remodeling induced by hypoxia. The phosphorylation levels of the PI3K, Akt, mTOR, PKC, and MAPK proteins were significantly up- or downregulated by RELM-β gene overexpression or silencing, respectively. Recombinant RELM-β protein increased the intracellular Ca2+ concentration in primary cultured hPASMCs and promoted hPASMC proliferation. The mitogenic effects of RELM-β on hPASMCs and the phosphorylation of PI3K, Akt, mTOR, PKC, and MAPK were suppressed by a Ca2+ inhibitor. Conclusions Our findings suggest that RELM-β acts as a cytokine-like growth factor in the development of HPH and that the effects of RELM-β are likely to be mediated by the Ca2+-dependent PI3K/Akt/mTOR and PKC/MAPK pathways.
Collapse
Affiliation(s)
- Heshen Tian
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China.,State Key Lab of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Lei Liu
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Ying Wu
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Ruiwen Wang
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Yongliang Jiang
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Ruicheng Hu
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Liming Zhu
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Linwei Li
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Yanyan Fang
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Chulan Yang
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Lianzhi Ji
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Guoyu Liu
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China
| | - Aiguo Dai
- Department of Respiratory Medicine & Department of Geriatric, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, People's Republic of China. .,Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, People's Republic of China.
| |
Collapse
|
20
|
Bryant AJ, Pham A, Gogoi H, Mitchell CR, Pais F, Jin L. The Third Man: DNA sensing as espionage in pulmonary vascular health and disease. Pulm Circ 2021; 11:2045894021996574. [PMID: 33738095 PMCID: PMC7934053 DOI: 10.1177/2045894021996574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 02/01/2021] [Indexed: 01/01/2023] Open
Abstract
For as long as nucleic acids have been utilized to vertically and horizontally transfer genetic material, living organisms have had to develop methods of recognizing cytosolic DNA as either pathogenic (microbial invasion) or physiologic (mitosis and cellular proliferation). Derangement in key signaling molecules involved in these pathways of DNA sensing result in a family of diseases labeled interferonopathies. An interferonopathy, characterized by constitutive expression of type I interferons, ultimately manifests as severe autoimmune disease at a young age. Afflicted patients present with a constellation of immune-mediated conditions, including primary lung manifestations such as pulmonary fibrosis and pulmonary hypertension. The latter condition is especially interesting in light of the known role that DNA damage plays in a variety of types of inherited and induced pulmonary hypertension, with free DNA detection elevated in the circulation of affected individuals. While little is known regarding the role of cytosolic DNA sensing in development of pulmonary vascular disease, exciting new research in the related fields of immunology and oncology potentially sheds light on future areas of fruitful exploration. As such, the goal of this review is to summarize the state of the field of nucleic acid sensing, extrapolating common shared pathways that parallel our knowledge of pulmonary hypertension, in a molecular and cell-specific manner. Principles of DNA sensing related to known pulmonary injury inducing stimuli are also evaluated, in addition to potential therapeutic targets. Finally, future directions in pulmonary hypertension research and treatments will be briefly discussed.
Collapse
Affiliation(s)
- Andrew J. Bryant
- University of Florida College of Medicine, Department of Medicine, Gainesville, FL, USA
| | - Ann Pham
- University of Florida College of Medicine, Department of Medicine, Gainesville, FL, USA
| | - Himanshu Gogoi
- University of Florida College of Medicine, Department of Medicine, Gainesville, FL, USA
| | - Carly R. Mitchell
- University of Florida College of Medicine, Department of Medicine, Gainesville, FL, USA
| | - Faye Pais
- University of Florida College of Medicine, Department of Medicine, Gainesville, FL, USA
| | - Lei Jin
- University of Florida College of Medicine, Department of Medicine, Gainesville, FL, USA
| |
Collapse
|
21
|
Golden JW, Cline CR, Zeng X, Garrison AR, Carey BD, Mucker EM, White LE, Shamblin JD, Brocato RL, Liu J, Babka AM, Rauch HB, Smith JM, Hollidge BS, Fitzpatrick C, Badger CV, Hooper JW. Human angiotensin-converting enzyme 2 transgenic mice infected with SARS-CoV-2 develop severe and fatal respiratory disease. JCI Insight 2020; 5:142032. [PMID: 32841215 PMCID: PMC7566707 DOI: 10.1172/jci.insight.142032] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/20/2020] [Indexed: 01/08/2023] Open
Abstract
The emergence of SARS-CoV-2 has created an international health crisis, and small animal models mirroring SARS-CoV-2 human disease are essential for medical countermeasure (MCM) development. Mice are refractory to SARS-CoV-2 infection owing to low-affinity binding to the murine angiotensin-converting enzyme 2 (ACE2) protein. Here, we evaluated the pathogenesis of SARS-CoV-2 in male and female mice expressing the human ACE2 gene under the control of the keratin 18 promoter (K18). In contrast to nontransgenic mice, intranasal exposure of K18-hACE2 animals to 2 different doses of SARS-CoV-2 resulted in acute disease, including weight loss, lung injury, brain infection, and lethality. Vasculitis was the most prominent finding in the lungs of infected mice. Transcriptomic analysis from lungs of infected animals showed increases in transcripts involved in lung injury and inflammatory cytokines. In the low-dose challenge groups, there was a survival advantage in the female mice, with 60% surviving infection, whereas all male mice succumbed to disease. Male mice that succumbed to disease had higher levels of inflammatory transcripts compared with female mice. To our knowledge, this is the first highly lethal murine infection model for SARS-CoV-2 and should be valuable for the study of SARS-CoV-2 pathogenesis and for the assessment of MCMs. A highly lethal murine infection model for SARS-CoV-2 using mice transgenic for the human ACE2 protein is described.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lauren E White
- Veterinary Medicine Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, USA
| | - Joshua D Shamblin
- Veterinary Medicine Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Lin Q, Johns RA. Resistin family proteins in pulmonary diseases. Am J Physiol Lung Cell Mol Physiol 2020; 319:L422-L434. [PMID: 32692581 DOI: 10.1152/ajplung.00040.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The family of resistin-like molecules (RELMs) consists of four members in rodents (RELMα/FIZZ1/HIMF, RELMβ/FIZZ2, Resistin/FIZZ3, and RELMγ/FIZZ4) and two members in humans (Resistin and RELMβ), all of which exhibit inflammation-regulating, chemokine, and growth factor properties. The importance of these cytokines in many aspects of physiology and pathophysiology, especially in cardiothoracic diseases, is rapidly evolving in the literature. In this review article, we attempt to summarize the contribution of RELM signaling to the initiation and progression of lung diseases, such as pulmonary hypertension, asthma/allergic airway inflammation, chronic obstructive pulmonary disease, fibrosis, cancers, infection, and other acute lung injuries. The potential of RELMs to be used as biomarkers or risk predictors of these diseases also will be discussed. Better understanding of RELM signaling in the pathogenesis of pulmonary diseases may offer novel targets or approaches for the development of therapeutics to treat or prevent a variety of inflammation, tissue remodeling, and fibrosis-related disorders in respiratory, cardiovascular, and other systems.
Collapse
Affiliation(s)
- Qing Lin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Roger A Johns
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
23
|
Krljanac B, Schubart C, Naumann R, Wirtz S, Culemann S, Krönke G, Voehringer D. RELMα-expressing macrophages protect against fatal lung damage and reduce parasite burden during helminth infection. Sci Immunol 2020; 4:4/35/eaau3814. [PMID: 31126996 DOI: 10.1126/sciimmunol.aau3814] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 01/08/2019] [Accepted: 04/26/2019] [Indexed: 12/15/2022]
Abstract
Alternatively activated macrophages (AAMs) can contribute to wound healing, regulation of glucose and fat metabolism, resolution of inflammation, and protective immunity against helminths. Their differentiation, tissue distribution, and effector functions are incompletely understood. Murine AAMs express high levels of resistin-like molecule (RELM) α, an effector protein with potent immunomodulatory functions. To visualize RELMα+ macrophages (MΦs) in vivo and evaluate their role in defense against helminths, we generated RELMα reporter/deleter mice. Infection with the helminth Nippostrongylus brasiliensis induced expansion of RELMα+ lung interstitial but not alveolar MΦs in a STAT6-dependent manner. RELMα+ MΦs were required for prevention of fatal lung damage during primary infection. Furthermore, protective immunity was lost upon specific deletion of RELMα+ MΦs during secondary infection. Thus, RELMα reporter/deleter mice reveal compartmentalization of AAMs in different tissues and demonstrate their critical role in resolution of severe lung inflammation and protection against migrating helminths.
Collapse
Affiliation(s)
- Branislav Krljanac
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Christoph Schubart
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Ronald Naumann
- Transgenic Core Facility, MPI of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Stefan Wirtz
- Department of Medicine 1-Gastroenterology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Stephan Culemann
- Department of Internal Medicine 3-Rheumatology and Immunology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3-Rheumatology and Immunology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany.
| |
Collapse
|
24
|
Lin Q, Fan C, Skinner JT, Hunter EN, Macdonald AA, Illei PB, Yamaji-Kegan K, Johns RA. RELMα Licenses Macrophages for Damage-Associated Molecular Pattern Activation to Instigate Pulmonary Vascular Remodeling. THE JOURNAL OF IMMUNOLOGY 2019; 203:2862-2871. [PMID: 31611261 DOI: 10.4049/jimmunol.1900535] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/23/2019] [Indexed: 01/21/2023]
Abstract
Pulmonary hypertension (PH) is a debilitating disease characterized by remodeling of the lung vasculature. In rodents, resistin-like molecule-α (RELMα, also known as HIMF or FIZZ1) can induce PH, but the signaling mechanisms are still unclear. In this study, we used human lung samples and a hypoxia-induced mouse model of PH. We found that the human homolog of RELMα, human (h) resistin, is upregulated in macrophage-like inflammatory cells from lung tissues of patients with idiopathic PH. Additionally, at PH onset in the mouse model, we observed RELMα-dependent lung accumulation of macrophages that expressed high levels of the key damage-associated molecular pattern (DAMP) molecule high-mobility group box 1 (HMGB1) and its receptor for advanced glycation end products (RAGE). In vitro, RELMα/hresistin-induced macrophage-specific HMGB1/RAGE expression and facilitated HMGB1 nucleus-to-cytoplasm translocation and extracellular secretion. Mechanistically, hresistin promoted HMGB1 posttranslational lysine acetylation by preserving the NAD+-dependent deacetylase sirtuin (Sirt) 1 in human macrophages. Notably, the hresistin-stimulated macrophages promoted apoptosis-resistant proliferation of human pulmonary artery smooth muscle cells in an HMGB1/RAGE-dependent manner. In the mouse model, RELMα also suppressed the Sirt1 signal in pulmonary macrophages in the early posthypoxic period. Notably, recruited macrophages in the lungs of these mice carried the RELMα binding partner Bruton tyrosine kinase (BTK). hResistin also mediated the migration of human macrophages by activating BTK in vitro. Collectively, these data reveal a vascular-immune cellular interaction in the early PH stage and suggest that targeting RELMα/DAMP-driven macrophages may offer a promising strategy to treat PH and other related vascular inflammatory diseases.
Collapse
Affiliation(s)
- Qing Lin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - Chunling Fan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - John T Skinner
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - Elizabeth N Hunter
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - Andrew A Macdonald
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - Peter B Illei
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Kazuyo Yamaji-Kegan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| | - Roger A Johns
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; and
| |
Collapse
|
25
|
Kassem KM, Ali M, Rhaleb NE. Interleukin 4: Its Role in Hypertension, Atherosclerosis, Valvular, and Nonvalvular Cardiovascular Diseases. J Cardiovasc Pharmacol Ther 2019; 25:7-14. [PMID: 31401864 DOI: 10.1177/1074248419868699] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hypertension is one of the major physiological risk factors for cardiovascular diseases, and it affects more than 1 billion adults worldwide, killing 9 million people every year according to World Health Organization. Also, hypertension is associated with increased risk of kidney disease and stroke. Studying the risk factors that contribute to the pathogenesis of hypertension is key to preventing and controlling hypertension. Numerous laboratories around to globe are very active pursuing research studies to delineate the factors, such as the role of immune system, which could contribute to hypertension. There are studies that were conducted on immune-deficient mice for which experimentally induced hypertension has been ameliorated. Thus, there are possibilities that immune reactivity could be associated with the development of certain type of hypertension. Furthermore, interleukin 4 has been associated with the development of pulmonary hypertension, which could lead to right ventricular remodeling. Also, the immune system is involved in valvular and nonvalvular cardiac remodeling. It has been demonstrated that there is a causative relationship between different interleukins and cardiac fibrosis.
Collapse
Affiliation(s)
- Kamal M Kassem
- Department of Internal Medicine, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Mahboob Ali
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nour-Eddine Rhaleb
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA.,Department of Physiology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
26
|
Nie Y, Zhang D, Qian F, Wu Y. Baccatin III ameliorates bleomycin-induced pulmonary fibrosis via suppression of TGF-β1 production and TGF-β1-induced fibroblast differentiation. Int Immunopharmacol 2019; 74:105696. [PMID: 31229901 DOI: 10.1016/j.intimp.2019.105696] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/12/2019] [Accepted: 06/12/2019] [Indexed: 12/25/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and generally lethal lung disease with a high mortality rate. Current therapeutic drugs exhibit limited efficacy but severe adverse effects. Paclitaxel has been identified to exert both anti-inflammatory and anti-fibrosis activity. Baccatin III (BAC), an important precursor of paclitaxel, has been identified as exhibiting immunomodulatory activity with decisively lower toxicity. However, its effects on pulmonary fibrosis remain unknown. In this study, the role of BAC in bleomycin (BLM)-induced pulmonary fibrosis and inflammation in mice was investigated in addition to elucidation of its mechanism of action. Our results demonstrated that administration of BAC in a dose-dependent manner reduced inflammatory infiltration, secretion of the pro-fibrotic mediator TGF-β1 and deposition of collagen and other components of the extracellular matrix (ECM), including alpha smooth muscle actin (α-SMA) and fibronectin. Administration of BAC to treat isolated macrophages stimulated with IL-13, known to activate macrophages, the principal source of TGF-β1, resulted in markedly reduced TGF-β1 expression from macrophages. The AKT/STAT6 signaling pathway was shown to be involved in this process. In addition, we have provided in vitro evidence that BAC inhibits TGF-β1-induced fibroblast differentiation via the Smad2/3 signaling pathway. Furthermore, intratracheal injection of rTGF-β1 significantly exacerbated the degree of fibrosis which was down-regulated by treatment with BAC. Taken together, our data suggest that BAC exerts a protective effect against lung fibrosis and may serve as a potential therapeutic strategy for IPF.
Collapse
Affiliation(s)
- Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, PR China
| | - Dan Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Feng Qian
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Yaxian Wu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, PR China.
| |
Collapse
|
27
|
Butrous G. Schistosome infection and its effect on pulmonary circulation. Glob Cardiol Sci Pract 2019; 2019:5. [PMID: 31024947 PMCID: PMC6472693 DOI: 10.21542/gcsp.2019.5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023] Open
Abstract
Schistosomiasis is the most common parasitic disease associated with pulmonary hypertension. It induces remodelling via complex inflammatory processes, which eventually produce the clinical manifestation of pulmonary hypertension. The pulmonary hypertension shows clinical signs and symptoms that are not distinguishable from other forms of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Ghazwan Butrous
- Professor of Cardiopulmonary Sciences, Medway School of Pharmacy, University of Kent, UK and University of Greenwich, Central Ave, Gillingham, Chatham ME4 4BF, Kent, UK
| |
Collapse
|
28
|
Willis GR, Fernandez-Gonzalez A, Reis M, Mitsialis SA, Kourembanas S. Macrophage Immunomodulation: The Gatekeeper for Mesenchymal Stem Cell Derived-Exosomes in Pulmonary Arterial Hypertension? Int J Mol Sci 2018; 19:ijms19092534. [PMID: 30150544 PMCID: PMC6164282 DOI: 10.3390/ijms19092534] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 12/13/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by remodeling of the pulmonary arteries, increased pulmonary infiltrates, loss of vascular cross-sectional area, and elevated pulmonary vascular resistance. Despite recent advances in the management of PAH, there is a pressing need for the development of new tools to effectively treat and reduce the risk of further complications. Dysregulated immunity underlies the development of PAH, and macrophages orchestrate both the initiation and resolution of pulmonary inflammation, thus, manipulation of lung macrophage function represents an attractive target for emerging immunomodulatory therapies, including cell-based approaches. Indeed, mesenchymal stem cell (MSC)-based therapies have shown promise, effectively modulating the macrophage fulcrum to favor an anti-inflammatory, pro-resolving phenotype, which is associated with both histological and functional benefits in preclinical models of pulmonary hypertension (PH). The complex interplay between immune system homeostasis and MSCs remains incompletely understood. Here, we highlight the importance of macrophage function in models of PH and summarize the development of MSC-based therapies, focusing on the significance of MSC exosomes (MEx) and the immunomodulatory and homeostatic mechanisms by which such therapies may afford their beneficial effects.
Collapse
Affiliation(s)
- Gareth R Willis
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Monica Reis
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - S Alex Mitsialis
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Stella Kourembanas
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
29
|
Pine GM, Batugedara HM, Nair MG. Here, there and everywhere: Resistin-like molecules in infection, inflammation, and metabolic disorders. Cytokine 2018; 110:442-451. [PMID: 29866514 DOI: 10.1016/j.cyto.2018.05.014] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 05/13/2018] [Accepted: 05/15/2018] [Indexed: 02/07/2023]
Abstract
The Resistin-Like Molecules (RELM) α, β, and γ and their namesake, resistin, share structural and sequence homology but exhibit significant diversity in expression and function within their mammalian host. RELM proteins are expressed in a wide range of diseases, such as: microbial infections (eg. bacterial and helminth), inflammatory diseases (eg. asthma, fibrosis) and metabolic disorders (eg. diabetes). While the expression pattern and molecular regulation of RELM proteins are well characterized, much controversy remains over their proposed functions, with evidence of host-protective and pathogenic roles. Moreover, the receptors for RELM proteins are unclear, although three receptors for resistin, decorin, adenylyl cyclase-associated protein 1 (CAP1), and Toll-like Receptor 4 (TLR4) have recently been proposed. In this review, we will first summarize the molecular regulation of the RELM gene family, including transcription regulation and tissue expression in humans and mouse disease models. Second, we will outline the function and receptor-mediated signaling associated with RELM proteins. Finally, we will discuss recent studies suggesting that, despite early misconceptions that these proteins are pathogenic, RELM proteins have a more nuanced and potentially beneficial role for the host in certain disease settings.
Collapse
Affiliation(s)
- Gabrielle M Pine
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, United States
| | - Hashini M Batugedara
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, United States
| | - Meera G Nair
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, United States.
| |
Collapse
|
30
|
LoPresti ST, Brown BN. Effect of Source Animal Age upon Macrophage Response to Extracellular Matrix Biomaterials. ACTA ACUST UNITED AC 2018; 1:57-66. [PMID: 30101208 DOI: 10.1016/j.regen.2018.03.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Extracellular matrix biomaterials have been shown to promote constructive remodeling in many preclinical and clinical applications. This response has been associated with the promotion of a timely switch from pro-inflammatory (M1) to anti-inflammatory (M2) macrophages. A previous study has shown that this beneficial response is lost when these biomaterials are derived from aged animals. This study examined the impact of small intestine submucosa (SIS) derived from 12, 26 and 52 week old pigs on the phenotype and function of bone marrow macrophages derived either from 2 or 18 month old mice. Results showed that 52 week old SIS promoted less iNOS in 2 month macrophages and Fizz1 expression in 2 and 18 month compared to 12 week SIS. Pro-inflammatory cytokine exposure to 52 week SIS-treated macrophages resulted in higher iNOS in 18 month macrophages and reduced MHC-II expression in 2 month macrophages, as well as reduced nitric oxide production in comparison to 12 week SIS. These results indicate that ECM derived from aged animals promotes an altered macrophage phenotype compared to young controls. This suggests that sourcing of ECM from young donors is important to preserve constructive remodeling outcomes of ECM biomaterials. Alteration of macrophage phenotype by aged ECM also raises the hypothesis that alterations in aged ECM may play a role in immune dysfunction in aged individuals.
Collapse
Affiliation(s)
- Samuel T LoPresti
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States.,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, United States
| | - Bryan N Brown
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States.,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, United States.,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, University of Pittsburgh, 300 Halket Street, Pittsburgh, PA 15213, United States
| |
Collapse
|
31
|
Li C, Liu P, Song R, Zhang Y, Lei S, Wu S. Immune cells and autoantibodies in pulmonary arterial hypertension. Acta Biochim Biophys Sin (Shanghai) 2017; 49:1047-1057. [PMID: 29036539 DOI: 10.1093/abbs/gmx095] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Indexed: 12/19/2022] Open
Abstract
Analyses of immunity in pulmonary arterial hypertension (PAH) support the notion that maladaptation of the immune response exists. Altered immunity is an increasingly recognized feature of PAH. Indeed, a delicate balance between immunity and tolerance exists and any disturbance may result in chronic inflammation or autoimmunity. This is suggested by infiltration of various immune cells (e.g. macrophages, T and B lymphocytes) in remodeled pulmonary vessels. In addition, several types of autoantibodies directed against antinuclear antigens, endothelial cells (ECs) and fibroblasts have been found in idiopathic and systemic sclerosis-associated PAH. These autoantibodies may play an important role in EC apoptosis and in the expression of cell adhesion molecules. This review article provides an overview of immunity pathways highlighting their potential roles in pulmonary vascular remodeling in PAH and the possibility of future targeted therapy.
Collapse
Affiliation(s)
- Cheng Li
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Pingping Liu
- Department of Emergency, Hunan Children's Hospital, Changsha, China
| | - Rong Song
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yiqing Zhang
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Si Lei
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Shangjie Wu
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
32
|
Hypoxia induced mitogenic factor (HIMF) triggers angiogenesis by increasing interleukin-18 production in myoblasts. Sci Rep 2017; 7:7393. [PMID: 28785068 PMCID: PMC5547156 DOI: 10.1038/s41598-017-07952-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 07/05/2017] [Indexed: 01/10/2023] Open
Abstract
Inflammatory myopathy is a rare autoimmune muscle disorder. Treatment typically focuses on skeletal muscle weakness or inflammation within muscle, as well as complications of respiratory failure secondary to respiratory muscle weakness. Impaired respiratory muscle function contributes to increased dyspnea and reduced exercise capacity in pulmonary hypertension (PH), a debilitating condition that has few treatment options. The initiation and progression of PH is associated with inflammation and inflammatory cell recruitment and it is established that hypoxia-induced mitogenic factor (HIMF, also known as resistin-like molecule α), activates macrophages in PH. However, the relationship between HIMF and inflammatory myoblasts remains unclear. This study investigated the signaling pathway involved in interleukin-18 (IL-18) expression and its relationship with HIMF in cultured myoblasts. We found that HIMF increased IL-18 production in myoblasts and that secreted IL-18 promoted tube formation of the endothelial progenitor cells. We used the mouse xenograft model and the chick chorioallantoic membrane assay to further explore the role of HIMF in inflammatory myoblasts and angiogenesis in vivo. Thus, our study focused on the mechanism by which HIMF mediates IL-18 expression in myoblasts through angiogenesis in vitro and in vivo. Our findings provide an insight into HIMF functioning in inflammatory myoblasts.
Collapse
|
33
|
Zeng X, Zhu L, Xiao R, Liu B, Sun M, Liu F, Hao Q, Lu Y, Zhang J, Li J, Wang T, Wei X, Hu Q. Hypoxia-Induced Mitogenic Factor Acts as a Nonclassical Ligand of Calcium-Sensing Receptor, Therapeutically Exploitable for Intermittent Hypoxia-Induced Pulmonary Hypertension. Hypertension 2017; 69:844-854. [PMID: 28348014 DOI: 10.1161/hypertensionaha.116.08743] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 12/03/2016] [Accepted: 02/26/2017] [Indexed: 11/16/2022]
Abstract
Hypoxia-induced mitogenic factor (HIMF) is an inflammatory cytokine playing important role(s) in the development of hypoxic pulmonary hypertension. The molecular target mediating HIMF-stimulated downstream events remains unclear. The coimmunoprecipitation screen identified extracellular calcium-sensing receptor (CaSR) as the binding partner for HIMF in pulmonary artery smooth muscle cells. The yeast 2-hybrid assay then revealed the binding of HIMF to the intracellular, not the extracellular, domain of extracellular CaSR. The binding of HIMF enhanced the activity of extracellular CaSR and mediated hypoxia-evoked proliferation of pulmonary artery smooth cells and the development of pulmonary vascular remodeling and pulmonary hypertension, all of which was specifically attenuated by a synthesized membrane-permeable peptide flanking the core amino acids of the intracellular binding domain of extracellular CaSR. Thus, HIMF induces pulmonary hypertension as a nonclassical ligand of extracellular CaSR, and the binding motif of extracellular CaSR can be therapeutically exploitable.
Collapse
Affiliation(s)
- Xianqin Zeng
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Liping Zhu
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Rui Xiao
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Bingxun Liu
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Mengxiang Sun
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fangbo Liu
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qiang Hao
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yankai Lu
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jiwei Zhang
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jiansha Li
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Tao Wang
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiang Wei
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qinghua Hu
- From the Department of Pathophysiology, School of Basic Medicine (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Q. Hu), Key Laboratory of Pulmonary Diseases of Ministry of Health (X.Z., L.Z., R.X., B.L., M.S., F.L., Q. Hao, Y.L., J.Z., J.L., T.W., Q. Hu), Department of Pathology, Tongji Hospital (Y.L., J.L.), Department of Pathology, Union Hospital (J.Z.), Department of Respiratory and Critical Care Medicine (T.W.), and Department of Cardiothoracic and Vascular Surgery, Tongji Hospital (X.W.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
34
|
Chen T, Yang C, Li M, Tan X. Alveolar Hypoxia-Induced Pulmonary Inflammation: From Local Initiation to Secondary Promotion by Activated Systemic Inflammation. J Vasc Res 2016; 53:317-329. [PMID: 27974708 DOI: 10.1159/000452800] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 10/23/2016] [Indexed: 11/19/2022] Open
Abstract
Pulmonary hypertension (PH) is a pathological condition with high mortality and morbidity. Hypoxic PH (HPH) is a common form of PH occurring mainly due to lung disease and/or hypoxia. Most causes of HPH are associated with persistent or intermittent alveolar hypoxia, including exposure to high altitude and chronic obstructive respiratory disease. Recent evidence suggests that inflammation is a critical step for HPH initiation and development. A detailed understanding of the initiation and progression of pulmonary inflammation would help in exploring potential clinical treatments for HPH. In this review, the mechanism for alveolar hypoxia-induced local lung inflammation and its progression are discussed as follows: (1) low alveolar PO2 levels activate resident lung cells, mainly the alveolar macrophages, which initiate pulmonary inflammation; (2) systemic inflammation is induced by alveolar hypoxia through alveolar macrophage activation; (3) monocytes are recruited into the pulmonary circulation by alveolar hypoxia-induced macrophage activation, which then contributes to the progression of pulmonary inflammation during the chronic phase of alveolar hypoxia, and (4) alveolar hypoxia-induced systemic inflammation contributes to the development of HPH. We hypothesize that a combination of alveolar hypoxia-induced local lung inflammation and the initiation of systemic inflammation ("second hit") is essential for HPH progression.
Collapse
Affiliation(s)
- Ting Chen
- Department of High Altitude Physiology and Biology, College of High Altitude Medicine, Third Military Medical University, Ministry of Education, Chongqing, China
| | | | | | | |
Collapse
|
35
|
Jackson IL, Zhang Y, Bentzen SM, Hu J, Zhang A, Vujaskovic Z. Pathophysiological mechanisms underlying phenotypic differences in pulmonary radioresponse. Sci Rep 2016; 6:36579. [PMID: 27845360 PMCID: PMC5109047 DOI: 10.1038/srep36579] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/12/2016] [Indexed: 12/25/2022] Open
Abstract
Differences in the pathogenesis of radiation-induced lung injury among murine strains offer a unique opportunity to elucidate the molecular mechanisms driving the divergence in tissue response from repair and recovery to organ failure. Here, we utilized two well-characterized murine models of radiation pneumonitis/fibrosis to compare and contrast differential gene expression in lungs 24 hours after exposure to a single dose of whole thorax lung irradiation sufficient to cause minor to major morbidity/mortality. Expression of 805 genes was altered as a general response to radiation; 42 genes were identified whose expression corresponded to the threshold for lethality. Three genes were discovered whose expression was altered within the lethal, but not the sublethal, dose range. Time-course analysis of the protein product of the most promising gene, resistin-like molecule alpha, demonstrated a significant difference in expression between radiosensitive versus radiotolerant strains, suggesting a unique role for this protein in acute lung injury.
Collapse
Affiliation(s)
- Isabel L Jackson
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yuji Zhang
- Division of Biostatistics and Bioinformatics, Department of Epidemiology &Public Health, and the Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Søren M Bentzen
- Division of Biostatistics and Bioinformatics, Department of Epidemiology &Public Health, and the Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jingping Hu
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Angel Zhang
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Zeljko Vujaskovic
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
36
|
Dehne N, Brüne B. Hypoxic inhibition of JMJD3 reduces H3K27me3 demethylation and induction of the STAT6 target gene CCL18. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1490-1501. [PMID: 27737800 DOI: 10.1016/j.bbagrm.2016.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/23/2016] [Accepted: 10/07/2016] [Indexed: 01/12/2023]
Abstract
Hypoxia, by activating transcription factors induces transcription of some genes but it also reduces mRNA synthesis by mechanisms that are poorly defined. Activation of human macrophages with interleukin (IL)-4 showed that up-regulation of some IL-4 target genes was reduced when macrophages were incubated at 1% oxygen. Hypoxia impaired induction of chemokine (C-C motif) ligand 18 (CCL18), although IL-4-induced DNA binding of the transcription factor STAT6 remained intact. In contrast, induction of serine peptidase inhibitor, Kunitz type (SPINT)2, another IL-4/STAT6 target gene, was not affected by hypoxia. The repressive histone mark histone 3 lysine 27 trimethylation (H3K27me3), known to prevent chromatin remodelling and transcription, was removed from the SPINT2 but not the CCL18 gene locus under hypoxia or dimethyloxalylglycine-treatment. The H3K27me3 demethylase JMJD3 was required for CCL18 gene induction but dispensable for induction of SPINT2. Our data indicate that hypoxic inhibition of JMJD3 activity reduces demethylation of H3K27me3, nucleosome removal, and hence induction of the STAT6 target gene CCL18, while induction of other STAT6-inducible genes such as SPINT2 remained unaffected by JMJD3. In contrast to mouse MΦ in human cells JMJD3 is not recruited by transcription factors like IRF4, KL4, or PPARγ to convey specificity in gene induction.
Collapse
Affiliation(s)
- Nathalie Dehne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany.
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
| |
Collapse
|
37
|
Dasgupta P, Dorsey NJ, Li J, Qi X, Smith EP, Yamaji-Kegan K, Keegan AD. The adaptor protein insulin receptor substrate 2 inhibits alternative macrophage activation and allergic lung inflammation. Sci Signal 2016; 9:ra63. [PMID: 27330190 DOI: 10.1126/scisignal.aad6724] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Insulin receptor substrate 2 (IRS2) is an adaptor protein that becomes tyrosine-phosphorylated in response to the cytokines interleukin-4 (IL-4) and IL-13, which results in activation of the phosphoinositide 3-kinase (PI3K)-Akt pathway. IL-4 and IL-13 contribute to allergic lung inflammation. To examine the role of IRS2 in allergic disease, we evaluated the responses of IRS2-deficient (IRS2(-/-)) mice. Unexpectedly, loss of IRS2 resulted in a substantial increase in the expression of a subset of genes associated with the generation of alternatively activated macrophages (AAMs) in response to IL-4 or IL-13 in vitro. AAMs secrete factors that enhance allergic responses and promote airway remodeling. Moreover, compared to IRS2(+/+) mice, IRS2(+/-) and IRS2(-/-) mice developed enhanced pulmonary inflammation, accumulated eosinophils and AAMs, and exhibited airway and vascular remodeling upon allergen stimulation, responses that partially depended on macrophage-intrinsic IRS2 signaling. Both in unstimulated and IL-4-stimulated macrophages, lack of IRS2 enhanced phosphorylation of Akt and ribosomal S6 protein. Thus, we identified a critical inhibitory loop downstream of IRS2, demonstrating an unanticipated and previously unrecognized role for IRS2 in suppressing allergic lung inflammation and remodeling.
Collapse
Affiliation(s)
- Preeta Dasgupta
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 West Baltimore Street Baltimore, MD 21201, USA. Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nicolas J Dorsey
- Medical Scientist Training Program, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jiaqi Li
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 West Baltimore Street Baltimore, MD 21201, USA
| | - Xiulan Qi
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 West Baltimore Street Baltimore, MD 21201, USA
| | - Elizabeth P Smith
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 West Baltimore Street Baltimore, MD 21201, USA
| | - Kazuyo Yamaji-Kegan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Achsah D Keegan
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 West Baltimore Street Baltimore, MD 21201, USA. Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA. Research and Development Service, U.S. Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD 21201, USA.
| |
Collapse
|
38
|
FIZZ1 Promotes Airway Remodeling in Asthma Through the PTEN Signaling Pathway. Inflammation 2016; 38:1464-72. [PMID: 25655389 DOI: 10.1007/s10753-015-0121-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aim of our study was to elucidate the function and signaling pathway of found in inflammatory zone 1 (FIZZ1) in airway remodeling in asthma. We used a mice model sensitized and challenged by ovalbumin (OVA) to evaluate the expression of FIZZ1, type I collagen, and fibronectin-1 in the airway in asthma. To investigate the signaling pathway regulated by FIZZ1, we treated a cultured murine lung epithelium cell-12 (MLE-12) with FIZZ1 recombination protein, silenced the expression of FIZZ1 with FIZZ1-shRNA in vitro, and then detected phosphorylated phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and expression of type I collagen and fibronectin-1 (FN-1) by Western blotting. In addition, we increased the expression of PTEN by PTEN plasmid transfection then detected the expression of type I collagen and fibronectin-1 in MLE-12 by Western blot analysis and immunofluorescence cytochemistry technology, respectively. First, the expression of FIZZ1, type I collagen, and fibronectin-1 was significantly elevated in the lungs of OVA-challenged mice compared with saline-treated control animals. Secondly, the phosphorylation of PTEN was decreased in MLE-12 treated with FIZZ1 recombination protein in vitro. On the contrary, the phosphorylation of PTEN was increased in MLE-12 cells transfected with FIZZ1-shRNA. Thirdly, results of the Western blot analysis and immunofluorescence cytochemistry showed that expression of type I collagen and fibronectin-1 was increased in cells treated with FIZZ1 recombination protein, while the levels of type I collagen and fibronectin-1 were significantly decreased in cells transfected with PTEN plasmid. FIZZ1 may be a critical cytokine in airway remodeling in asthma. This study indicates that targeting FIZZ1 and/or PTEN may be a new therapeutic strategy for asthma.
Collapse
|
39
|
Jiang R, Zhao C, Wang X, Wang S, Sun X, Tian Y, Song W. Resistin-Like Molecule-β Promotes Invasion and Migration of Gastric Carcinoma Cells. Med Sci Monit 2016; 22:937-42. [PMID: 27001185 PMCID: PMC4806584 DOI: 10.12659/msm.895598] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Resistin-like molecule-β (RELMβ) is a novel secretory protein from intestinal goblet cells and participates in epithelial differentiation, tumor occurrence, and immune response. RELMβ is absent in normal gastric mucosa but is abundantly expressed in gastric carcinoma tissues, and is correlated with tumor invasion and metastasis. Epithelial-mesenchymal transition (EMT) is an important mechanism governing tumor cell invasion. This study thus investigated the modulation of RELMβ in gastric cancer metastasis and its correlation with EMT. Material/Methods We used RELMβ-low expression AGS cell line of gastric cancer and normal mucosa cell line GES1 as in vitro models, on which RELMβ0-expressing vector was transfected. The invasion and migration of cells were quantified by Transwell assay. EMT-related protein including E-cadherin, N-cadherin, Snail, and Vimentin were detected by Western blotting in transfected AGS cells. Results RELMβ transfection significantly potentiated invasion and migration abilities of AGS cells, whose RELMβ protein level was significantly elevated compared to those in untransfected AGS or GES1 cells. After RELMβ transfection, EMT-related proteins, including N-cadherin, Snail, and Vimentin levels, were elevated, but E-cadherin expression was depressed. Conclusions RELMβ-overexpression can facilitate invasion and migration of gastric carcinoma cells and it increases the expression of EMT-related proteins, such as N-cadherin, Snail, Vimentin, but decreases E-cadherin level, thus promoting the progression of EMT.
Collapse
Affiliation(s)
- Rui Jiang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Chunming Zhao
- Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Xinyu Wang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Shengxi Wang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Xiaogang Sun
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Yang Tian
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Wei Song
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
40
|
Johns RA, Takimoto E, Meuchel LW, Elsaigh E, Zhang A, Heller NM, Semenza GL, Yamaji-Kegan K. Hypoxia-Inducible Factor 1α Is a Critical Downstream Mediator for Hypoxia-Induced Mitogenic Factor (FIZZ1/RELMα)-Induced Pulmonary Hypertension. Arterioscler Thromb Vasc Biol 2015; 36:134-44. [PMID: 26586659 DOI: 10.1161/atvbaha.115.306710] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 11/05/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Pulmonary hypertension (PH) is characterized by progressive elevation of pulmonary vascular resistance, right ventricular failure, and ultimately death. We have shown that in rodents, hypoxia-induced mitogenic factor (HIMF; also known as FIZZ1 or resistin-like molecule-β) causes PH by initiating lung vascular inflammation. We hypothesized that hypoxia-inducible factor-1 (HIF-1) is a critical downstream signal mediator of HIMF during PH development. APPROACH AND RESULTS In this study, we compared the degree of HIMF-induced pulmonary vascular remodeling and PH development in wild-type (HIF-1α(+/+)) and HIF-1α heterozygous null (HIF-1α(+/-)) mice. HIMF-induced PH was significantly diminished in HIF-1α(+/-) mice and was accompanied by a dysregulated vascular endothelial growth factor-A-vascular endothelial growth factor receptor 2 pathway. HIF-1α was critical for bone marrow-derived cell migration and vascular tube formation in response to HIMF. Furthermore, HIMF and its human homolog, resistin-like molecule-β, significantly increased interleukin (IL)-6 in macrophages and lung resident cells through a mechanism dependent on HIF-1α and, at least to some extent, on nuclear factor κB. CONCLUSIONS Our results suggest that HIF-1α is a critical downstream transcription factor for HIMF-induced pulmonary vascular remodeling and PH development. Importantly, both HIMF and human resistin-like molecule-β significantly increased IL-6 in lung resident cells and increased perivascular accumulation of IL-6-expressing macrophages in the lungs of mice. These data suggest that HIMF can induce HIF-1, vascular endothelial growth factor-A, and interleukin-6, which are critical mediators of both hypoxic inflammation and PH pathophysiology.
Collapse
Affiliation(s)
- Roger A Johns
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Eiki Takimoto
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Lucas W Meuchel
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Esra Elsaigh
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Ailan Zhang
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Nicola M Heller
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Gregg L Semenza
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Kazuyo Yamaji-Kegan
- Department of Anesthesiology and Critical Care Medicine (R.A.J., L.W.M., E.E., A.Z., N.M.H., K.Y.-K.), the Division of Cardiology (E.T.), and Vascular Program, Institute for Cell Engineering, Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry (G.L.S.), McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD.
| |
Collapse
|
41
|
Understanding the Mysterious M2 Macrophage through Activation Markers and Effector Mechanisms. Mediators Inflamm 2015; 2015:816460. [PMID: 26089604 PMCID: PMC4452191 DOI: 10.1155/2015/816460] [Citation(s) in RCA: 1239] [Impact Index Per Article: 123.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/30/2015] [Indexed: 11/17/2022] Open
Abstract
The alternatively activated or M2 macrophages are immune cells with high phenotypic heterogeneity and are governing functions at the interface of immunity, tissue homeostasis, metabolism, and endocrine signaling. Today the M2 macrophages are identified based on the expression pattern of a set of M2 markers. These markers are transmembrane glycoproteins, scavenger receptors, enzymes, growth factors, hormones, cytokines, and cytokine receptors with diverse and often yet unexplored functions. This review discusses whether these M2 markers can be reliably used to identify M2 macrophages and define their functional subdivisions. Also, it provides an update on the novel signals of the tissue environment and the neuroendocrine system which shape the M2 activation. The possible evolutionary roots of the M2 macrophage functions are also discussed.
Collapse
|
42
|
Zhu R, Bi LQ, Wu SL, Li L, Kong H, Xie WP, Wang H, Meng ZL. Iptakalim attenuates hypoxia-induced pulmonary arterial hypertension in rats by endothelial function protection. Mol Med Rep 2015; 12:2945-52. [PMID: 25936382 DOI: 10.3892/mmr.2015.3695] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 01/09/2015] [Indexed: 11/05/2022] Open
Abstract
The present study aimed to investigate the protective effects of iptakalim, an adenosine triphosphate (ATP)-sensitive potassium channel opener, on the inflammation of the pulmonary artery and endothelial cell injury in a hypoxia-induced pulmonary arterial hypertension (PAH) rat model. Ninety-six Sprague-Dawley rats were placed into normobaric hypoxia chambers for four weeks and were treated with iptakalim (1.5 mg/kg/day) or saline for 28 days. The right ventricle systolic pressures (RVSP) were measured and small pulmonary arterial morphological alterations were analyzed with hematoxylin and eosin staining. Enzyme-linked immunosorbent assay (ELISA) was performed to analyze the content of interleukin (IL)-1β and IL-10. Immunohistochemical analysis for ED1(+) monocytes was performed to detect the inflammatory cells surrounding the pulmonary arterioles. Western blot analysis was performed to analyze the expression levels of platelet endothelial cell adhesion molecule-1 (PECAM-1) and endothelial nitric oxide synthase (eNOS) in the lung tissue. Alterations in small pulmonary arteriole morphology and the ultrastructure of pulmonary arterial endothelial cells were observed via light and transmission electron microscopy, respectively. Iptakalim significantly attenuated the increase in mean pulmonary artery pressure, RVSP, right ventricle to left ventricle plus septum ratio and small pulmonary artery wall remodeling in hypoxia-induced PAH rats. Iptakalim also prevented an increase in IL-1β and a decrease in IL-10 in the peripheral blood and lung tissue, and alleviated inflammatory cell infiltration in hypoxia-induced PAH rats. Furthermore, iptakalim enhanced PECAM-1 and eNOS expression and prevented the endothelial cell injury induced by hypoxic stimuli. Iptakalim suppressed the pulmonary arteriole and systemic inflammatory responses and protected against the endothelial damage associated with the upregulation of PECAM-1 and eNOS, suggesting that iptakalim may represent a potential therapeutic agent for PAH.
Collapse
Affiliation(s)
- Rong Zhu
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Li-Qing Bi
- Geriatric Intensive Care Unit, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Su-Ling Wu
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Lan Li
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hui Kong
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wei-Ping Xie
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hong Wang
- Department of Respiratory Medicine, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zi-Li Meng
- Department of Respiratory Medicine, The Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| |
Collapse
|
43
|
Broytman O, Braun RK, Morgan BJ, Pegelow DF, Hsu PN, Mei LS, Koya AK, Eldridge M, Teodorescu M. Effects of Chronic Intermittent Hypoxia on Allergen-Induced Airway Inflammation in Rats. Am J Respir Cell Mol Biol 2015; 52:162-70. [DOI: 10.1165/rcmb.2014-0213oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
44
|
Park SH, Chen WC, Esmaeil N, Lucas B, Marsh LM, Reibman J, Grunig G. Interleukin 13- and interleukin 17A-induced pulmonary hypertension phenotype due to inhalation of antigen and fine particles from air pollution. Pulm Circ 2015; 4:654-68. [PMID: 25610601 DOI: 10.1086/678511] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 04/10/2014] [Indexed: 12/12/2022] Open
Abstract
Pulmonary hypertension has a marked detrimental effect on quality of life and life expectancy. In a mouse model of antigen-induced pulmonary arterial remodeling, we have recently shown that coexposure to urban ambient particulate matter (PM) significantly increased the thickening of the pulmonary arteries and also resulted in significantly increased right ventricular systolic pressures. Here we interrogate the mechanism and show that combined neutralization of interleukin 13 (IL-13) and IL-17A significantly ameliorated the increase in right ventricular systolic pressure, the circumferential muscularization of pulmonary arteries, and the molecular change in the right ventricle. Surprisingly, our data revealed a protective role of IL-17A for the antigen- and PM-induced severe thickening of pulmonary arteries. This protection was due to the inhibition of the effects of IL-13, which drove this response, and the expression of metalloelastase and resistin-like molecule α. However, the latter was redundant for the arterial thickening response. Anti-IL-13 exacerbated airway neutrophilia, which was due to a resulting excess effect of IL-17A, confirming concurrent cross inhibition of IL-13- and IL-17A-dependent responses in the lungs of animals exposed to antigen and PM. Our experiments also identified IL-13/IL-17A-independent molecular reprogramming in the lungs induced by exposure to antigen and PM, which indicates a risk for arterial remodeling and protection from arterial constriction. Our study points to IL-13- and IL-17A-coinduced inflammation as a new template for biomarkers and therapeutic targeting for the management of immune response-induced pulmonary hypertension.
Collapse
Affiliation(s)
- Sung-Hyun Park
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Wen-Chi Chen
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Nafiseh Esmaeil
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA ; Current affiliation: Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Benjamin Lucas
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Leigh M Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Joan Reibman
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA ; Pulmonary Medicine, Department of Medicine, New York University School of Medicine, New York, New York, USA
| | - Gabriele Grunig
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA ; Pulmonary Medicine, Department of Medicine, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
45
|
Lee MR, Shim D, Yoon J, Jang HS, Oh SW, Suh SH, Choi JH, Oh GT. Retnla overexpression attenuates allergic inflammation of the airway. PLoS One 2014; 9:e112666. [PMID: 25415454 PMCID: PMC4240542 DOI: 10.1371/journal.pone.0112666] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 10/10/2014] [Indexed: 02/07/2023] Open
Abstract
Resistin-like molecule alpha (Retnla), also known as ‘Found in inflammatory zone 1’, is a secreted protein that has been found in bronchoalveolar lavage (BAL) fluid of ovalbumin (OVA)-induced asthmatic mice and plays a role as a regulator of T helper (Th)2-driven inflammation. However, the role of Retnla in the progress of Th2-driven airway inflammation is not yet clear. To better understand the function of Retnla in Th2-driven airway inflammation, we generated Retnla-overexpressing (Retnla-Tg) mice. Retnla-Tg mice showed increased expression of Retnla protein in BAL fluid and airway epithelial cells. Retnla overexpression itself did not induce any alteration in lung histology or lung function compared to non-Tg controls. However, OVA-sensitized/challenged Retnla-Tg mice had decreased numbers of cells in BAL and inflammatory cells accumulating in the lung. They also showed a reduction in mucus production in the airway epithelium, concomitant with a decreased Muc5ac level. These results were accompanied by reduced levels of Th2 cytokines, including interleukin (IL)-4, IL-5, and IL-13, with no effect on levels of OVA-specific immunoglobulin isotypes. Furthermore, phosphorylation of ERK was markedly reduced in the lungs of OVA-challenged Retnla-Tg mice. Taken together, these results indicates that Retnla protects against Th2-mediated inflammation in an experimental mouse model of asthma, suggesting that therapeutic approaches to enhance the production of Retnla or Retnla-like molecules could be valuable for preventing allergic lung inflammation.
Collapse
Affiliation(s)
- Mi-Ran Lee
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Dahee Shim
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Jihye Yoon
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Hyung Seok Jang
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Se-Woong Oh
- Yuhan Research Institute, Yuhan Corporation, Gongse-Dong, Giheung-Gu, Yongin-Si, Gyeonggi-Do, Republic of Korea
| | - Suk Hyo Suh
- Department of Physiology Medical School, Ewha Womans University, Seoul, Republic of Korea
| | - Jae-Hoon Choi
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
- * E-mail: (JHC); (GTO)
| | - Goo Taeg Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
- * E-mail: (JHC); (GTO)
| |
Collapse
|
46
|
Pugliese SC, Poth JM, Fini MA, Olschewski A, El Kasmi KC, Stenmark KR. The role of inflammation in hypoxic pulmonary hypertension: from cellular mechanisms to clinical phenotypes. Am J Physiol Lung Cell Mol Physiol 2014; 308:L229-52. [PMID: 25416383 DOI: 10.1152/ajplung.00238.2014] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hypoxic pulmonary hypertension (PH) comprises a heterogeneous group of diseases sharing the common feature of chronic hypoxia-induced pulmonary vascular remodeling. The disease is usually characterized by mild to moderate pulmonary vascular remodeling that is largely thought to be reversible compared with the progressive irreversible disease seen in World Health Organization (WHO) group I disease. However, in these patients, the presence of PH significantly worsens morbidity and mortality. In addition, a small subset of patients with hypoxic PH develop "out-of-proportion" severe pulmonary hypertension characterized by pulmonary vascular remodeling that is irreversible and similar to that in WHO group I disease. In all cases of hypoxia-related vascular remodeling and PH, inflammation, particularly persistent inflammation, is thought to play a role. This review focuses on the effects of hypoxia on pulmonary vascular cells and the signaling pathways involved in the initiation and perpetuation of vascular inflammation, especially as they relate to vascular remodeling and transition to chronic irreversible PH. We hypothesize that the combination of hypoxia and local tissue factors/cytokines ("second hit") antagonizes tissue homeostatic cellular interactions between mesenchymal cells (fibroblasts and/or smooth muscle cells) and macrophages and arrests these cells in an epigenetically locked and permanently activated proremodeling and proinflammatory phenotype. This aberrant cellular cross-talk between mesenchymal cells and macrophages promotes transition to chronic nonresolving inflammation and vascular remodeling, perpetuating PH. A better understanding of these signaling pathways may lead to the development of specific therapeutic targets, as none are currently available for WHO group III disease.
Collapse
Affiliation(s)
- Steven C Pugliese
- Developmental Lung Biology, Cardiovascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatrics-Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado;
| | - Jens M Poth
- Developmental Lung Biology, Cardiovascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatrics-Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Mehdi A Fini
- Developmental Lung Biology, Cardiovascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatrics-Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria; and
| | - Karim C El Kasmi
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, University of Colorado Denver, School of Medicine, Anschutz Medical Campus, Aurora, Colorado
| | - Kurt R Stenmark
- Developmental Lung Biology, Cardiovascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatrics-Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
47
|
El Kasmi KC, Pugliese SC, Riddle SR, Poth JM, Anderson AL, Frid MG, Li M, Pullamsetti SS, Savai R, Nagel MA, Fini MA, Graham BB, Tuder RM, Friedman JE, Eltzschig HK, Sokol RJ, Stenmark KR. Adventitial fibroblasts induce a distinct proinflammatory/profibrotic macrophage phenotype in pulmonary hypertension. THE JOURNAL OF IMMUNOLOGY 2014; 193:597-609. [PMID: 24928992 DOI: 10.4049/jimmunol.1303048] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Macrophage accumulation is not only a characteristic hallmark but is also a critical component of pulmonary artery remodeling associated with pulmonary hypertension (PH). However, the cellular and molecular mechanisms that drive vascular macrophage activation and their functional phenotype remain poorly defined. Using multiple levels of in vivo (bovine and rat models of hypoxia-induced PH, together with human tissue samples) and in vitro (primary mouse, rat, and bovine macrophages, human monocytes, and primary human and bovine fibroblasts) approaches, we observed that adventitial fibroblasts derived from hypertensive pulmonary arteries (bovine and human) regulate macrophage activation. These fibroblasts activate macrophages through paracrine IL-6 and STAT3, HIF1, and C/EBPβ signaling to drive expression of genes previously implicated in chronic inflammation, tissue remodeling, and PH. This distinct fibroblast-activated macrophage phenotype was independent of IL-4/IL-13-STAT6 and TLR-MyD88 signaling. We found that genetic STAT3 haplodeficiency in macrophages attenuated macrophage activation, complete STAT3 deficiency increased macrophage activation through compensatory upregulation of STAT1 signaling, and deficiency in C/EBPβ or HIF1 attenuated fibroblast-driven macrophage activation. These findings challenge the current paradigm of IL-4/IL-13-STAT6-mediated alternative macrophage activation as the sole driver of vascular remodeling in PH, and uncover a cross-talk between adventitial fibroblasts and macrophages in which paracrine IL-6-activated STAT3, HIF1α, and C/EBPβ signaling are critical for macrophage activation and polarization. Thus, targeting IL-6 signaling in macrophages by completely inhibiting C/EBPβ or HIF1α or by partially inhibiting STAT3 may hold therapeutic value for treatment of PH and other inflammatory conditions characterized by increased IL-6 and absent IL-4/IL-13 signaling.
Collapse
Affiliation(s)
- Karim C El Kasmi
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, CO 80045;
| | - Steven C Pugliese
- Division of Critical Care Medicine/Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Suzette R Riddle
- Division of Critical Care Medicine/Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Jens M Poth
- Division of Critical Care Medicine/Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Aimee L Anderson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Maria G Frid
- Division of Critical Care Medicine/Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Min Li
- Division of Critical Care Medicine/Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Soni S Pullamsetti
- Department of Lung Development and Remodeling, Max-Planck Institute for Heart and Lung Research, University of Giessen and Marburg Lung Center, German Center for Lung Research, D-61231 Bad Nauheim, Germany
| | - Rajkumar Savai
- Department of Lung Development and Remodeling, Max-Planck Institute for Heart and Lung Research, University of Giessen and Marburg Lung Center, German Center for Lung Research, D-61231 Bad Nauheim, Germany
| | - Maria A Nagel
- Department of Neurology, University of Colorado Denver, School of Medicine, Aurora, CO 80045
| | - Mehdi A Fini
- Division of Critical Care Medicine/Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Brian B Graham
- Program in Translational Lung Research, Department of Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Rubin M Tuder
- Program in Translational Lung Research, Department of Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Jacob E Friedman
- Division of Biochemistry and Molecular Genetics, Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, CO 80045; and
| | - Holger K Eltzschig
- Department of Anesthesiology, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Ronald J Sokol
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, School of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Kurt R Stenmark
- Division of Critical Care Medicine/Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, School of Medicine, University of Colorado Denver, Aurora, CO 80045;
| |
Collapse
|
48
|
Li W, Holsinger RMD, Kruse CA, Flügel A, Graeber MB. The potential for genetically altered microglia to influence glioma treatment. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2014; 12:750-62. [PMID: 24047526 DOI: 10.2174/18715273113126660171] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/06/2012] [Accepted: 12/06/2012] [Indexed: 01/06/2023]
Abstract
Diffuse and unstoppable infiltration of brain and spinal cord tissue by neoplastic glial cells is the single most important therapeutic problem posed by the common glioma group of tumors: astrocytoma, oligoastrocytoma, oligodendroglioma, their malignant variants and glioblastoma. These neoplasms account for more than two thirds of all malignant central nervous system tumors. However, most glioma research focuses on an examination of the tumor cells rather than on host-specific, tumor micro-environmental cells and factors. This can explain why existing diffuse glioma therapies fail and why these tumors have remained incurable. Thus, there is a great need for innovation. We describe a novel strategy for the development of a more effective treatment of diffuse glioma. Our approach centers on gaining control over the behavior of the microglia, the defense cells of the CNS, which are manipulated by malignant glioma and support its growth. Armoring microglia against the influences from glioma is one of our research goals. We further discuss how microglia precursors may be genetically enhanced to track down infiltrating glioma cells.
Collapse
Affiliation(s)
- W Li
- Brain and Mind Research Institute, The University of Sydney, Camperdown, NSW, Australia.
| | | | | | | | | |
Collapse
|
49
|
Yamaji-Kegan K, Takimoto E, Zhang A, Weiner NC, Meuchel LW, Berger AE, Cheadle C, Johns RA. Hypoxia-induced mitogenic factor (FIZZ1/RELMα) induces endothelial cell apoptosis and subsequent interleukin-4-dependent pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2014; 306:L1090-103. [PMID: 24793164 DOI: 10.1152/ajplung.00279.2013] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pulmonary hypertension (PH) is characterized by elevated pulmonary artery pressure that leads to progressive right heart failure and ultimately death. Injury to endothelium and consequent wound repair cascades have been suggested to trigger pulmonary vascular remodeling, such as that observed during PH. The relationship between injury to endothelium and disease pathogenesis in this disorder remains poorly understood. We and others have shown that, in mice, hypoxia-induced mitogenic factor (HIMF, also known as FIZZ1 or RELMα) plays a critical role in the pathogenesis of lung inflammation and the development of PH. In this study, we dissected the mechanism by which HIMF and its human homolog resistin (hRETN) induce pulmonary endothelial cell (EC) apoptosis and subsequent lung inflammation-mediated PH, which exhibits many of the hallmarks of the human disease. Systemic administration of HIMF caused increases in EC apoptosis and interleukin (IL)-4-dependent vascular inflammatory marker expression in mouse lung during the early inflammation phase. In vitro, HIMF, hRETN, and IL-4 activated pulmonary microvascular ECs (PMVECs) by increasing angiopoietin-2 expression and induced PMVEC apoptosis. In addition, the conditioned medium from hRETN-treated ECs had elevated levels of endothelin-1 and caused significant increases in pulmonary vascular smooth muscle cell proliferation. Last, HIMF treatment caused development of PH that was characterized by pulmonary vascular remodeling and right heart failure in wild-type mice but not in IL-4 knockout mice. These data suggest that HIMF contributes to activation of vascular inflammation at least in part by inducing EC apoptosis in the lung. These events lead to subsequent PH.
Collapse
Affiliation(s)
- Kazuyo Yamaji-Kegan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland;
| | - Eiki Takimoto
- Division of Cardiology, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Ailan Zhang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Noah C Weiner
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Lucas W Meuchel
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Alan E Berger
- Divison of Allergy and Clinical Immunology, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Chris Cheadle
- Divison of Allergy and Clinical Immunology, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Roger A Johns
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| |
Collapse
|
50
|
Swain SD, Siemsen DW, Pullen RR, Han S. CD4+ T cells and IFN-γ are required for the development of Pneumocystis-associated pulmonary hypertension. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 184:483-93. [PMID: 24361497 DOI: 10.1016/j.ajpath.2013.10.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 09/11/2013] [Accepted: 10/23/2013] [Indexed: 12/24/2022]
Abstract
Pulmonary hypertension (PH) is a disease of diverse etiology. Although primary PH can develop in the absence of prior disease, PH more commonly develops in conjunction with other pulmonary pathologies. We previously reported a mouse model in which PH occurs as a sequela of Pneumocystis infection in the context of transient CD4 depletion. Here, we report that instead of the expected Th2 pathways, the Th1 cytokine IFN-γ is essential for the development of PH, as wild-type mice developed PH but IFN-γ knockout mice did not. Because gene expression analysis showed few strain differences that were not immune-function related, we focused on those responses as potential pathologic mechanisms. In addition to dependence on IFN-γ, we found that when CD4 cells were continuously depleted, but infection was limited by antibiotic treatment, PH did not occur, confirming that CD4 T cells are required for PH development. Also, although CD8 T-cells are implicated in the pathology of Pneumocystis pneumonia, they did not have a role in the onset of PH. Finally, we found differences in immune cell phenotypes that correlated with PH, including elevated CD204 expression in lung CD11c(+) cells, but their role remains unclear. Overall, we demonstrate that a transient, localized, immune response requiring IFN-γ and CD4-T cells can disrupt pulmonary vascular function and promote lingering PH.
Collapse
Affiliation(s)
- Steve D Swain
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana.
| | - Dan W Siemsen
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana
| | - Rebecca R Pullen
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana
| | - Soo Han
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, Montana
| |
Collapse
|