1
|
Su X, Sun Y, Dai A. New insights into pulmonary arterial hypertension: interaction between PANoptosis and perivascular inflammatory responses. Apoptosis 2025:10.1007/s10495-025-02086-0. [PMID: 39979525 DOI: 10.1007/s10495-025-02086-0] [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] [Accepted: 01/24/2025] [Indexed: 02/22/2025]
Abstract
Pulmonary arterial hypertension (PAH) is a heterogeneous disease characterized by various etiologies, with pulmonary vascular remodeling recognized as a main pathological change. Currently, it is widely accepted that vascular remodeling is closely associated with abnormal pulmonary vascular cell death and perivascular inflammation. The simultaneous activation of various pulmonary vascular cell death leads to immune cell adhesion and inflammatory mediator releases; And in turn, the inflammatory response may also trigger cell death and jointly promote the progression of vascular remodeling. Recently, PANoptosis has been identified as a phenomenon that describes the simultaneous activation and interaction of multiple forms of programmed cell death (PCD). Therefore, the relationship between PANoptosis and inflammation in PAH warrants further investigation. This review examines the mechanisms underlying apoptosis, necroptosis, pyroptosis, and inflammatory responses in PAH, with a focus on PANoptosis and its interactions with inflammation. And it aims to elucidate the significance of this emerging form of cell death and inflammation in the pathophysiology of PAH and to explore its potential as a therapeutic target.
Collapse
Affiliation(s)
- Xianli Su
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Yinhui Sun
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Aiguo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China.
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China.
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China.
| |
Collapse
|
2
|
Baglivo I, Quaranta VN, Dragonieri S, Colantuono S, Menzella F, Selvaggio D, Carpagnano GE, Caruso C. The New Paradigm: The Role of Proteins and Triggers in the Evolution of Allergic Asthma. Int J Mol Sci 2024; 25:5747. [PMID: 38891935 PMCID: PMC11171572 DOI: 10.3390/ijms25115747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Epithelial barrier damage plays a central role in the development and maintenance of allergic inflammation. Rises in the epithelial barrier permeability of airways alter tissue homeostasis and allow the penetration of allergens and other external agents. Different factors contribute to barrier impairment, such as eosinophilic infiltration and allergen protease action-eosinophilic cationic proteins' effects and allergens' proteolytic activity both contribute significantly to epithelial damage. In the airways, allergen proteases degrade the epithelial junctional proteins, allowing allergen penetration and its uptake by dendritic cells. This increase in allergen-immune system interaction induces the release of alarmins and the activation of type 2 inflammatory pathways, causing or worsening the main symptoms at the skin, bowel, and respiratory levels. We aim to highlight the molecular mechanisms underlying allergenic protease-induced epithelial barrier damage and the role of immune response in allergic asthma onset, maintenance, and progression. Moreover, we will explore potential clinical and radiological biomarkers of airway remodeling in allergic asthma patients.
Collapse
Affiliation(s)
- Ilaria Baglivo
- Centro Malattie Apparato Digerente (CEMAD) Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Vitaliano Nicola Quaranta
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Silvano Dragonieri
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Stefania Colantuono
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Francesco Menzella
- Pulmonology Unit, S. Valentino Hospital-AULSS2 Marca Trevigiana, 31100 Treviso, Italy
| | - David Selvaggio
- UOS di Malattie dell’Apparato Respiratorio Ospedale Cristo Re, 00167 Roma, Italy
| | - Giovanna Elisiana Carpagnano
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, Section of Respiratory Disease, University “Aldo Moro” of Bari, 70121 Bari, Italy (S.D.)
| | - Cristiano Caruso
- Unità Operativa Semplice Dipartimentale Day Hospital (UOSD DH) Medicina Interna e Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| |
Collapse
|
3
|
Zhang YS, Chen YQ. Dysfunctional regulatory T cell: May be an obstacle to immunotherapy in cardiovascular diseases. Biomed Pharmacother 2024; 173:116359. [PMID: 38430633 DOI: 10.1016/j.biopha.2024.116359] [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: 12/06/2023] [Revised: 02/18/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024] Open
Abstract
Inflammatory responses are linked to cardiovascular diseases (CVDs) in various forms. Tregs, members of CD4+ T cells, play important roles in regulating immune system and suppressing inflammatory response, thus contributing to maintaining immune homeostasis. However, Tregs exert their powerful suppressive function relying on the stable phenotype and function. The stability of Tregs primarily depends on the FOXP3 (Forkhead box P3) expression and epigenetic regulation. Although Tregs are quite stable under physiological conditions, prolonged exposure to inflammatory cues, Tregs may lose suppressive function and require proinflammatory phenotype, namely plastic Tregs or ex-Tregs. There are extensive researches have established the beneficial role of Tregs in CVDs. Nevertheless, the potential risks of dysfunctional Tregs lack deep research. Anti-inflammatory and immunological modulation have been hotspots in the treatment of CVDs. Tregs are appealing because of their crucial role in resolving inflammation and promoting tissue repair. If alleviating inflammatory response through modulating Tregs could be a new therapeutic strategy for CVDs, the next step to consider is how to prevent the formation of dysfunctional Tregs or reverse detrimental Tregs to normal phenotype.
Collapse
Affiliation(s)
- Yu-Sha Zhang
- Department of Cardiology, the Second Xiangya Hospital, Central South University, Hunan, China
| | - Ya-Qin Chen
- Department of Cardiology, the Second Xiangya Hospital, Central South University, Hunan, China.
| |
Collapse
|
4
|
Thoreau B, Mouthon L. Pulmonary arterial hypertension associated with connective tissue diseases (CTD-PAH): Recent and advanced data. Autoimmun Rev 2024; 23:103506. [PMID: 38135175 DOI: 10.1016/j.autrev.2023.103506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 12/17/2023] [Indexed: 12/24/2023]
Abstract
Pulmonary arterial hypertension (PAH), corresponding to group 1 of pulmonary hypertension classification, is a rare disease with a major prognostic impact on morbidity and mortality. PAH can be either primary in idiopathic and heritable forms or secondary to other conditions including connective tissue diseases (CTD-PAH). Within CTD-PAH, the leading cause of PAH is systemic sclerosis (SSc) in Western countries, whereas systemic lupus erythematosus (SLE) and mixed connective tissue disease (MCTD) are predominantly associated with PAH in Asia. Although many advances have been made during the last two decades regarding classification, definition early screening and risk stratification and therapeutic aspects with initial combination treatment, the specificities of CTD-PAH are not yet clear. In this manuscript, we review recent literature data regarding the updated definition and classification of PAH, pathogenesis, epidemiology, detection, prognosis and treatment of CTD-PAH.
Collapse
Affiliation(s)
- Benjamin Thoreau
- Department of Internal Medicine, Referral Center for Rare Autoimmune and Systemic Diseases, AP-HP.Centre, Université Paris Cité, Hôpital Cochin, 27, rue du Faubourg Saint-Jacques, 75679 Cedex 14 Paris, France; Université Paris Cité, F-75006 Paris, France; INSERM U1016, Cochin Institute, CNRS UMR 8104, Université Paris Cité, Paris, France.
| | - Luc Mouthon
- Department of Internal Medicine, Referral Center for Rare Autoimmune and Systemic Diseases, AP-HP.Centre, Université Paris Cité, Hôpital Cochin, 27, rue du Faubourg Saint-Jacques, 75679 Cedex 14 Paris, France; Université Paris Cité, F-75006 Paris, France; INSERM U1016, Cochin Institute, CNRS UMR 8104, Université Paris Cité, Paris, France
| |
Collapse
|
5
|
Soh WT, Zhang J, Hollenberg MD, Vliagoftis H, Rothenberg ME, Sokol CL, Robinson C, Jacquet A. Protease allergens as initiators-regulators of allergic inflammation. Allergy 2023; 78:1148-1168. [PMID: 36794967 PMCID: PMC10159943 DOI: 10.1111/all.15678] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 02/05/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
Tremendous progress in the last few years has been made to explain how seemingly harmless environmental proteins from different origins can induce potent Th2-biased inflammatory responses. Convergent findings have shown the key roles of allergens displaying proteolytic activity in the initiation and progression of the allergic response. Through their propensity to activate IgE-independent inflammatory pathways, certain allergenic proteases are now considered as initiators for sensitization to themselves and to non-protease allergens. The protease allergens degrade junctional proteins of keratinocytes or airway epithelium to facilitate allergen delivery across the epithelial barrier and their subsequent uptake by antigen-presenting cells. Epithelial injuries mediated by these proteases together with their sensing by protease-activated receptors (PARs) elicit potent inflammatory responses resulting in the release of pro-Th2 cytokines (IL-6, IL-25, IL-1β, TSLP) and danger-associated molecular patterns (DAMPs; IL-33, ATP, uric acid). Recently, protease allergens were shown to cleave the protease sensor domain of IL-33 to produce a super-active form of the alarmin. At the same time, proteolytic cleavage of fibrinogen can trigger TLR4 signaling, and cleavage of various cell surface receptors further shape the Th2 polarization. Remarkably, the sensing of protease allergens by nociceptive neurons can represent a primary step in the development of the allergic response. The goal of this review is to highlight the multiple innate immune mechanisms triggered by protease allergens that converge to initiate the allergic response.
Collapse
Affiliation(s)
- Wai Tuck Soh
- Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Jihui Zhang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Morley D. Hollenberg
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Harissios Vliagoftis
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine & Dentistry, and Alberta Respiratory Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Marc E. Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Caroline L. Sokol
- Division of Rheumatology, Allergy and Immunology, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Clive Robinson
- Institute for Infection and Immunity, St George’s University of London, London, UK
| | - Alain Jacquet
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
6
|
Huang L, Li H, Huang S, Wang S, Liu Q, Luo L, Gan S, Fu G, Zou P, Chen G, Wu Z. Notopterol Attenuates Monocrotaline-Induced Pulmonary Arterial Hypertension in Rat. Front Cardiovasc Med 2022; 9:859422. [PMID: 35722110 PMCID: PMC9203832 DOI: 10.3389/fcvm.2022.859422] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Current targeted pulmonary arterial hypertension (PAH) therapies have improved lung hemodynamics, cardiac function, and quality of life; however, none of these have reversed the ongoing remodeling of blood vessels. Considering notopterol, a linear furocoumarin extracted from the root of traditional Chinese medicine Qiang-Huo (Notopterygium incisum), had shown the antiproliferative and anti-inflammatory properties in previous studies, we hypothesized that it could play a role in ameliorating PAH. Methods In vivo, we conducted monocrotaline (MCT) induced PAH rats and treated them with notopterol for 3 weeks. Then, the rats were examined by echocardiography and RV catheterization. The heart and lung specimens were harvested for the detection of gross examination, histological examination and expression of inflammatory molecules. In vitro, human pulmonary arterial smooth muscle cells (HPASMCs) were treated with notopterol after hypoxia; then, cell proliferation was assessed by cell counting kit-8 and Edu assay, and cell migration was detected by wound healing assays. Results We found that notopterol improved mortality rate and RV function while reducing right ventricular systolic pressure in MCT-induced PAH rats. Furthermore, notopterol reduced right ventricular hypertrophy and fibrosis, and it also eased pulmonary vascular remodeling and MCT-induced muscularization. In addition, notopterol attenuated the pro-inflammatory factor (IL-1β, IL-6) and PCNA in the lungs of PAH rats. For the cultured HPASMCs subjected to hypoxia, we found that notopterol can inhibit the proliferation and migration of HPASMCs. Conclusion Our studies show that notopterol exerts anti-inflammatory and anti-proliferative effects in the pulmonary arteries, which may contribute to prevention of PAH.
Collapse
Affiliation(s)
- Lin Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Huayang Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Suiqing Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shunjun Wang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Quan Liu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Li Luo
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuangjiao Gan
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guangguo Fu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - PeiYun Zou
- GuangZhou Janus Biotechnology Co. Ltd., Guangzhou, China
| | - Guangxian Chen
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Guangxian Chen
| | - Zhongkai Wu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Zhongkai Wu
| |
Collapse
|
7
|
The role of immune cells in pulmonary hypertension: Focusing on macrophages. Hum Immunol 2021; 83:153-163. [PMID: 34844784 DOI: 10.1016/j.humimm.2021.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/14/2021] [Accepted: 11/15/2021] [Indexed: 01/06/2023]
Abstract
Pulmonary hypertension (PH) is a life-threatening pathological state with elevated pulmonary arterial pressure, resulting in right ventricular failure and heart functional failure. Analyses of human samples and rodent models of pH support the infiltration of various immune cells, including neutrophils, mast cells, dendritic cells, B-cells, T-cells, and natural killer cells, to the lungs and pulmonary perivascular regions and their involvement in the PH development. There is evidence that macrophages are presented in the pulmonary lesions of pH patients as first-line myeloid leucocytes. Macrophage accumulation and presence, both M1 and M2 phenotypes, is a distinctive hallmark of pH which plays a pivotal role in pulmonary artery remodeling through various cellular and molecular interactions and mechanisms, including CCL2 and CX3CL1 chemokines, adventitial fibroblasts, glucocorticoid-regulated kinase 1 (SGK1), crosstalk with other immune cells, leukotriene B4 (LTB4), bone morphogenetic protein receptor 2 (BMPR2), macrophage migration inhibitory factor (MIF), and thrombospondin-1 (TSP-1). In this paper, we reviewed the molecular mechanisms and the role of immune cells and responses are involved in PH development. We also summarized the polarization of macrophages in response to different stimuli and their pathological role and their infiltration in the lung of pH patients and animal models.
Collapse
|
8
|
Karoor V, Strassheim D, Sullivan T, Verin A, Umapathy NS, Dempsey EC, Frank DN, Stenmark KR, Gerasimovskaya E. The Short-Chain Fatty Acid Butyrate Attenuates Pulmonary Vascular Remodeling and Inflammation in Hypoxia-Induced Pulmonary Hypertension. Int J Mol Sci 2021; 22:9916. [PMID: 34576081 PMCID: PMC8467617 DOI: 10.3390/ijms22189916] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/01/2021] [Accepted: 09/10/2021] [Indexed: 12/30/2022] Open
Abstract
Pulmonary hypertension (PH) is a progressive cardiovascular disorder in which local vascular inflammation leads to increased pulmonary vascular remodeling and ultimately to right heart failure. The HDAC inhibitor butyrate, a product of microbial fermentation, is protective in inflammatory intestinal diseases, but little is known regarding its effect on extraintestinal diseases, such as PH. In this study, we tested the hypothesis that butyrate is protective in a Sprague-Dawley (SD) rat model of hypoxic PH. Treatment with butyrate (220 mg/kg intake) prevented hypoxia-induced right ventricular hypertrophy (RVH), hypoxia-induced increases in right ventricular systolic pressure (RVSP), pulmonary vascular remodeling, and permeability. A reversal effect of butyrate (2200 mg/kg intake) was observed on elevated RVH. Butyrate treatment also increased the acetylation of histone H3, 25-34 kDa, and 34-50 kDa proteins in the total lung lysates of butyrate-treated animals. In addition, butyrate decreased hypoxia-induced accumulation of alveolar (mostly CD68+) and interstitial (CD68+ and CD163+) lung macrophages. Analysis of cytokine profiles in lung tissue lysates showed a hypoxia-induced upregulation of TIMP-1, CINC-1, and Fractalkine and downregulation of soluble ICAM (sICAM). The expression of Fractalkine and VEGFα, but not CINC-1, TIMP-1, and sICAM was downregulated by butyrate. In rat microvascular endothelial cells (RMVEC), butyrate (1 mM, 2 and 24 h) exhibited a protective effect against TNFα- and LPS-induced barrier disruption. Butyrate (1 mM, 24 h) also upregulated tight junctional proteins (occludin, cingulin, claudin-1) and increased the acetylation of histone H3 but not α-tubulin. These findings provide evidence of the protective effect of butyrate on hypoxic PH and suggest its potential use as a complementary treatment for PH and other cardiovascular diseases.
Collapse
Affiliation(s)
- Vijaya Karoor
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (V.K.); (D.S.); (T.S.); (E.C.D.); (K.R.S.)
| | - Derek Strassheim
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (V.K.); (D.S.); (T.S.); (E.C.D.); (K.R.S.)
| | - Timothy Sullivan
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (V.K.); (D.S.); (T.S.); (E.C.D.); (K.R.S.)
| | - Alexander Verin
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; (A.V.); (N.S.U.)
| | - Nagavedi S. Umapathy
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; (A.V.); (N.S.U.)
- Center for Blood Disorders, Augusta University, Augusta, GA 30912, USA
| | - Edward C. Dempsey
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (V.K.); (D.S.); (T.S.); (E.C.D.); (K.R.S.)
- Rocky Mountain Regional VA Center, Aurora, CO 80045, USA
| | - Daniel N. Frank
- Division of Infectious Diseases, Department of Medicine, University of Colorado Denver, Denver, CO 80204, USA;
| | - Kurt R. Stenmark
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (V.K.); (D.S.); (T.S.); (E.C.D.); (K.R.S.)
- Division of Critical Care Medicine, Department of Pediatrics, University of Colorado Denver, Denver, CO 80204, USA
| | - Evgenia Gerasimovskaya
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (V.K.); (D.S.); (T.S.); (E.C.D.); (K.R.S.)
- Division of Critical Care Medicine, Department of Pediatrics, University of Colorado Denver, Denver, CO 80204, USA
| |
Collapse
|
9
|
Anderson LJ, Jadhao SJ, Paden CR, Tong S. Functional Features of the Respiratory Syncytial Virus G Protein. Viruses 2021; 13:1214. [PMID: 34372490 PMCID: PMC8310105 DOI: 10.3390/v13071214] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/28/2021] [Accepted: 06/18/2021] [Indexed: 12/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of serious lower respiratory tract infections in children <5 years of age worldwide and repeated infections throughout life leading to serious disease in the elderly and persons with compromised immune, cardiac, and pulmonary systems. The disease burden has made it a high priority for vaccine and antiviral drug development but without success except for immune prophylaxis for certain young infants. Two RSV proteins are associated with protection, F and G, and F is most often pursued for vaccine and antiviral drug development. Several features of the G protein suggest it could also be an important to vaccine or antiviral drug target design. We review features of G that effect biology of infection, the host immune response, and disease associated with infection. Though it is not clear how to fit these together into an integrated picture, it is clear that G mediates cell surface binding and facilitates cellular infection, modulates host responses that affect both immunity and disease, and its CX3C aa motif contributes to many of these effects. These features of G and the ability to block the effects with antibody, suggest G has substantial potential in vaccine and antiviral drug design.
Collapse
Affiliation(s)
- Larry J. Anderson
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA;
| | - Samadhan J. Jadhao
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA;
| | - Clinton R. Paden
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30322, USA; (C.R.P.); (S.T.)
| | - Suxiang Tong
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30322, USA; (C.R.P.); (S.T.)
| |
Collapse
|
10
|
Liang S, Desai AA, Black SM, Tang H. Cytokines, Chemokines, and Inflammation in Pulmonary Arterial Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:275-303. [PMID: 33788198 DOI: 10.1007/978-3-030-63046-1_15] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
According to the World Symposium Pulmonary Hypertension (WSPH) classification, pulmonary hypertension (PH) is classified into five categories based on etiology. Among them, Group 1 pulmonary arterial hypertension (PAH) disorders are rare but progressive and often, fatal despite multiple approved treatments. Elevated pulmonary arterial pressure in patients with WSPH Group 1 PAH is mainly caused by increased pulmonary vascular resistance (PVR), due primarily to sustained pulmonary vasoconstriction and excessive obliterative pulmonary vascular remodeling. Growing evidence indicates that inflammation plays a critical role in the development of pulmonary vascular remodeling associated with PAH. While the role of auto-immunity is unclear, infiltration of inflammatory cells in and around vascular lesions, including T- and B-cells, dendritic cells, macrophages, and mast cells have been observed in PAH patients. Serum and plasma levels of chemokines, cytokines, and autoantibodies are also increased in PAH patients; some of these circulating molecules are correlated with disease severity and survival. Preclinical experiments have reported a key role of the inflammation in PAH pathophysiology in vivo. Importantly, anti-inflammatory and immunosuppressive agents have further exhibited therapeutic effects. The present chapter reviews published experimental and clinical evidence highlighting the canonical role of inflammation in the pathogenesis of PAH and as a major target for the development of anti-inflammatory therapies in patients with PAH.
Collapse
Affiliation(s)
- Shuxin Liang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.,State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ankit A Desai
- Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Stephen M Black
- Division of Translational and Regenerative Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Haiyang Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China. .,State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
11
|
Gewehr DM, Salgueiro GR, de Noronha L, Kubrusly FB, Kubrusly LF, Coltro GA, Preto PC, Bertoldi ADS, Vieira HI. Plexiform Lesions in an Experimental Model of Monocrotalin-Induced Pulmonary Arterial Hypertension. Arq Bras Cardiol 2020; 115:480-490. [PMID: 33027370 PMCID: PMC9363102 DOI: 10.36660/abc.20190306] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/27/2019] [Accepted: 08/18/2019] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND The monocrotaline (MCT)-induced pulmonary arterial hypertension model is one of the most reproduced today, presenting as a limitation the absence of plexiform lesions, typical manifestations of the severe disease in humans. OBJECTIVE To evaluate the severity of MCT-induced pulmonary arteriopathy by pathological findings of lung and heart tissue samples, clinical course and 37-day survival. METHODS Fifty male Wistar rats were divided into one of the four groups - control (CG) (n = 10) and three intervention (MCT) groups. The MCT groups received intraperitoneal injection (60 mg/kg) of MCT and remained exposed to the substance for 15 days (G15, n = 10), 30 days (G30, n = 10) and 37 days (G37, n = 20). At the end of each period, the animals were sacrificed, and pulmonary and cardiac tissues were collected for anatomopathological and morphometric analysis. The Kruskal-Wallis test was used, considering a level of significance of 5%. RESULTS In the lungs of MCT animals, lesions related to pulmonary arteriopathy were found, including muscularization of the arterioles, hypertrophy of the middle layer and concentric neointimal lesions. Complex lesions were observed in MCT groups, described as plexiform and plexiform-like lesions. Right ventricular hypertrophy was evidenced by increased thickness and diameter of the cardiomyocytes and a significant increase in the right ventricular wall thickness (p <0.0000). CONCLUSION The MCT model was able to generate moderate-severe pulmonary arteriopathy associated with secondary right ventricular hypertrophy. The 37-day survival rate was 50%. To our knowledge, this study was the first to note the presence of complex vascular lesions, similar to those observed in patients with severe pulmonary arterial hypertension, in an isolated MCT model. (Arq Bras Cardiol. 2020; 115(3):480-490).
Collapse
Affiliation(s)
- Douglas Mesadri Gewehr
- Faculdade Evangélica Mackenzie do ParanáCuritibaPRBrasilFaculdade Evangélica Mackenzie do Paraná (FEMPAR), Curitiba, PR - Brasil
- Instituto Denton Cooley de Pesquisa, Ciência e TecnologiaCuritibaPRBrasilInstituto Denton Cooley de Pesquisa, Ciência e Tecnologia (IDC),Curitiba, PR - Brasil
| | - Gabriela Rodrigues Salgueiro
- Faculdade Evangélica Mackenzie do ParanáCuritibaPRBrasilFaculdade Evangélica Mackenzie do Paraná (FEMPAR), Curitiba, PR - Brasil
- Instituto Denton Cooley de Pesquisa, Ciência e TecnologiaCuritibaPRBrasilInstituto Denton Cooley de Pesquisa, Ciência e Tecnologia (IDC),Curitiba, PR - Brasil
| | - Lucia de Noronha
- Pontifícia Universidade Católica do ParanáDepartamento de MedicinaCuritibaPRBrasilPontifícia Universidade Católica do Paraná Departamento de Medicina, Curitiba, PR - Brasil
| | - Fernando Bermudez Kubrusly
- Instituto Denton Cooley de Pesquisa, Ciência e TecnologiaCuritibaPRBrasilInstituto Denton Cooley de Pesquisa, Ciência e Tecnologia (IDC),Curitiba, PR - Brasil
- Instituto do Coração de CuritibaCuritibaPRBrasilInstituto do Coração de Curitiba (InCor Curitiba), Curitiba, PR - Brasil
| | - Luiz Fernando Kubrusly
- Faculdade Evangélica Mackenzie do ParanáCuritibaPRBrasilFaculdade Evangélica Mackenzie do Paraná (FEMPAR), Curitiba, PR - Brasil
- Instituto Denton Cooley de Pesquisa, Ciência e TecnologiaCuritibaPRBrasilInstituto Denton Cooley de Pesquisa, Ciência e Tecnologia (IDC),Curitiba, PR - Brasil
- Instituto do Coração de CuritibaCuritibaPRBrasilInstituto do Coração de Curitiba (InCor Curitiba), Curitiba, PR - Brasil
| | - Gabriel Antonio Coltro
- Faculdade Evangélica Mackenzie do ParanáCuritibaPRBrasilFaculdade Evangélica Mackenzie do Paraná (FEMPAR), Curitiba, PR - Brasil
- Instituto Denton Cooley de Pesquisa, Ciência e TecnologiaCuritibaPRBrasilInstituto Denton Cooley de Pesquisa, Ciência e Tecnologia (IDC),Curitiba, PR - Brasil
| | - Paola Cardoso Preto
- Pontifícia Universidade Católica do ParanáDepartamento de MedicinaCuritibaPRBrasilPontifícia Universidade Católica do Paraná Departamento de Medicina, Curitiba, PR - Brasil
| | - Andressa de Souza Bertoldi
- Instituto Denton Cooley de Pesquisa, Ciência e TecnologiaCuritibaPRBrasilInstituto Denton Cooley de Pesquisa, Ciência e Tecnologia (IDC),Curitiba, PR - Brasil
- Centro de Estudos e Pesquisa em Emergências Médicas e Terapia IntensivaCuritibaPRBrasilCentro de Estudos e Pesquisa em Emergências Médicas e Terapia Intensiva (CEPETI), Curitiba, PR - Brasil
| | - Heloisa Iacomo Vieira
- Hospital Universitário Evangélico MackenzieCuritibaPRBrasilHospital Universitário Evangélico Mackenzie (HUEM), Curitiba, PR – Brasil
| |
Collapse
|
12
|
Siamwala JH, Zhao A, Barthel H, Pagano FS, Gilbert RJ, Rounds S. Adaptive and innate immune mechanisms in cardiac fibrosis complicating pulmonary arterial hypertension. Physiol Rep 2020; 8:e14532. [PMID: 32786064 PMCID: PMC7422804 DOI: 10.14814/phy2.14532] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a syndrome diagnosed by increased mean pulmonary artery (PA) pressure and resistance and normal pulmonary capillary wedge pressure. PAH is characterized pathologically by distal pulmonary artery remodeling, increased pulmonary vascular resistance, and plexiform lesions (PLs). Right ventricular fibrosis and hypertrophy, leading to right ventricular failure, are the main determinants of mortality in PAH. Recent work suggests that right ventricular fibrosis results from resident cardiac fibroblast activation and conversion to myofibroblasts, leading to replacement of contractile cardiomyocytes with nondistensible tissue incapable of conductivity or contractility. However, the origins, triggers, and consequences of myofibroblast expansion and its pathophysiological relationship with PAH are unclear. Recent advances indicate that signals generated by adaptive and innate immune cells may play a role in right ventricular fibrosis and remodeling. This review summarizes recent insights into the mechanisms by which adaptive and innate immune signals participate in the transition of cardiac fibroblasts to activated myofibroblasts and highlights the existing gaps of knowledge as relates to the development of right ventricular fibrosis.
Collapse
Affiliation(s)
- Jamila H. Siamwala
- Department of Molecular PharmacologyPhysiology and BiotechnologyBrown UniversityProvidenceRIUSA
- Warren Alpert Medical School of Brown UniversityProvidence VA Medical CenterProvidenceRIUSA
| | - Alexander Zhao
- Department of Molecular PharmacologyPhysiology and BiotechnologyBrown UniversityProvidenceRIUSA
| | - Haley Barthel
- Department of Molecular PharmacologyPhysiology and BiotechnologyBrown UniversityProvidenceRIUSA
| | - Francesco S. Pagano
- Department of Molecular PharmacologyPhysiology and BiotechnologyBrown UniversityProvidenceRIUSA
| | - Richard J. Gilbert
- Ocean State Research InstituteProvidence VA Medical CenterProvidenceRIUSA
| | - Sharon Rounds
- Warren Alpert Medical School of Brown UniversityProvidence VA Medical CenterProvidenceRIUSA
- Department of MedicineDivision of PulmonaryCritical Care and SleepWarren Alpert Medical School of Brown UniversityProvidenceRIUSA
| |
Collapse
|
13
|
Kim K, Choi JH. Involvement of immune responses in pulmonary arterial hypertension; lessons from rodent models. Lab Anim Res 2019; 35:22. [PMID: 32257910 PMCID: PMC7081631 DOI: 10.1186/s42826-019-0021-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/09/2019] [Indexed: 12/18/2022] Open
Abstract
Pulmonary hypertension (PH) is a pathological state with sustained elevation of pulmonary artery (PA) pressure. Since the pathogenesis of PH is mostly irreversible, the disease often comes up with poor prognosis. Pulmonary arterioles are affected by deteriorative changes, such as development of occlusive lesions of thickening of arterial walls. Such processes increase the pulmonary arterial pressure thus lead to consequent injuries such as right ventricle failure. Proliferation, or resistance to apoptosis of pulmonary artery smooth muscle cells (PASMC) and fibroblasts, are characteristic changes observed in the PA in pulmonary arterial hypertension (PAH) patients. PAH can either occur idiopathically or come with other diseases. Emerging evidences suggest that pro-inflammatory processes are closely related to the development of PAH. Therefore, it is inferred that immune cells could be the key factors in PAH development. In this review, we summarize the way how each types of immune cells participate in PAH. We would also like to list the current rodent models used for PAH study.
Collapse
Affiliation(s)
- Kibyeong Kim
- Department of Life Science, College of Natural Sciences, Research Institute of Natural Sciences Hanyang University, Seoul, Republic of Korea
| | - Jae-Hoon Choi
- Department of Life Science, College of Natural Sciences, Research Institute of Natural Sciences Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
14
|
Mamazhakypov A, Viswanathan G, Lawrie A, Schermuly RT, Rajagopal S. The role of chemokines and chemokine receptors in pulmonary arterial hypertension. Br J Pharmacol 2019; 178:72-89. [PMID: 31399998 DOI: 10.1111/bph.14826] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by progressive pulmonary artery remodelling leading to increased right ventricular pressure overload, which results in right heart failure and premature death. Inflammation plays a central role in the development of PAH, and the recruitment and function of immune cells are tightly regulated by chemotactic cytokines called chemokines. A number of studies have shown that the development and progression of PAH are associated with the dysregulated expression of several chemokines and chemokine receptors in the pulmonary vasculature. Moreover, some chemokines are differentially regulated in the pressure-overloaded right ventricle. Recent studies have tested the efficacy of pharmacological agents targeting several chemokines and chemokine receptors for their effects on the development of PAH, suggesting that these receptors could serve as useful therapeutic targets. In this review, we provide recent insights into the role of chemokines and chemokine receptors in PAH and RV remodelling and the opportunities and roadblocks in targeting them. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.1/issuetoc.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Gayathri Viswanathan
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Allan Lawrie
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Ralph Theo Schermuly
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Giessen, Germany
| | - Sudarshan Rajagopal
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
15
|
Boyoglu-Barnum S, Chirkova T, Anderson LJ. Biology of Infection and Disease Pathogenesis to Guide RSV Vaccine Development. Front Immunol 2019; 10:1675. [PMID: 31402910 PMCID: PMC6677153 DOI: 10.3389/fimmu.2019.01675] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/04/2019] [Indexed: 12/21/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of severe lower respiratory tract disease in young children and a substantial contributor to respiratory tract disease throughout life and as such a high priority for vaccine development. However, after nearly 60 years of research no vaccine is yet available. The challenges to developing an RSV vaccine include the young age, 2-4 months of age, for the peak of disease, the enhanced RSV disease associated with the first RSV vaccine, formalin-inactivated RSV with an alum adjuvant (FI-RSV), and difficulty achieving protection as illustrated by repeat infections with disease that occur throughout life. Understanding the biology of infection and disease pathogenesis has and will continue to guide vaccine development. In this paper, we review the roles that RSV proteins play in the biology of infection and disease pathogenesis and the corresponding contribution to live attenuated and subunit RSV vaccines. Each of RSV's 11 proteins are in the design of one or more vaccines. The G protein's contribution to disease pathogenesis through altering host immune responses as well as its role in the biology of infection suggest it can make a unique contribution to an RSV vaccine, both live attenuated and subunit vaccines. One of G's potential unique contributions to a vaccine is the potential for anti-G immunity to have an anti-inflammatory effect independent of virus replication. Though an anti-viral effect is essential to an effective RSV vaccine, it is important to remember that the goal of a vaccine is to prevent disease. Thus, other effects of the infection, such as G's alteration of the host immune response may provide opportunities to induce responses that block this effect and improve an RSV vaccine. Keeping in mind the goal of a vaccine is to prevent disease and not virus replication may help identify new strategies for other vaccine challenges, such as improving influenza vaccines and developing HIV vaccines.
Collapse
Affiliation(s)
| | - Tatiana Chirkova
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Larry J. Anderson
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, GA, United States
| |
Collapse
|
16
|
Abid S, Marcos E, Parpaleix A, Amsellem V, Breau M, Houssaini A, Vienney N, Lefevre M, Derumeaux G, Evans S, Hubeau C, Delcroix M, Quarck R, Adnot S, Lipskaia L. CCR2/CCR5-mediated macrophage–smooth muscle cell crosstalk in pulmonary hypertension. Eur Respir J 2019; 54:13993003.02308-2018. [DOI: 10.1183/13993003.02308-2018] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 06/27/2019] [Indexed: 11/05/2022]
Abstract
Macrophages are major players in the pathogenesis of pulmonary arterial hypertension (PAH).To investigate whether lung macrophages and pulmonary-artery smooth muscle cells (PASMCs) collaborate to stimulate PASMC growth and whether the CCL2-CCR2 and CCL5-CCR5 pathways inhibited macrophage–PASMC interactions and PAH development, we used human CCR5-knock-in mice and PASMCs from patients with PAH and controls.Conditioned media from murine M1 or M2 macrophages stimulated PASMC growth. This effect was markedly amplified with conditioned media from M2 macrophage/PASMC co-cultures. CCR2, CCR5, CCL2 and CCL5 were upregulated in macrophage/PASMC co-cultures. Compared to inhibiting either receptor, dual CCR2 and CCR5 inhibition more strongly attenuated the growth-promoting effect of conditioned media from M2-macrophage/PASMC co-cultures. Deleting either CCR2 or CCR5 in macrophages or PASMCs attenuated the growth response. In mice with hypoxia- or SUGEN/hypoxia-induced PH, targeting both CCR2 and CCR5 prevented or reversed PH more efficiently than targeting either receptor alone. Patients with PAH exhibited CCR2 and CCR5 upregulation in PASMCs and perivascular macrophages compared to controls. The PASMC growth-promoting effect of conditioned media from M2-macrophage/PASMC co-cultures was greater when PASMCs from PAH patients were used in the co-cultures or as the target cells and was dependent on CCR2 and CCR5. PASMC migration toward M2-macrophages was greater with PASMCs from PAH patients and was attenuated by blocking CCR2 and CCR5.CCR2 and CCR5 are required for collaboration between macrophages and PASMCs to initiate and amplify PASMC migration and proliferation during PAH development. Dual targeting of CCR2 and CCR5 may hold promise for treating human PAH.
Collapse
|
17
|
Xiao B, Liu F, Lu JC, Chen F, Pei WN, Yang XC. IGF-1 deletion affects renal sympathetic nerve activity, left ventricular dysfunction, and renal function in DOCA-salt hypertensive mice. Physiol Res 2019; 68:209-217. [PMID: 30628826 DOI: 10.33549/physiolres.933918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
To determine the influence of IGF-1 deletion on renal sympathetic nerve activity (RSNA), left ventricular dysfunction, and renal function in deoxycorticosterone acetate (DOCA)-salt hypertensive mice. The DOCA-salt hypertensive mice models were constructed and the experiment was classified into WT (Wild-type mice) +sham, LID (Liver-specific IGF-1 deficient mice) + sham, WT + DOCA, and LID+ DOCA groups. Enzyme-linked immunosorbent assay (ELISA) was used to detect the serum IGF-1 levels in mice. The plasma norepinephrine (NE), urine protein, urea nitrogen and creatinine, as well as RSNA were measured. Echocardiography was performed to assess left ventricular dysfunction, and HE staining to observe the pathological changes in renal tissue of mice. DOCA-salt induction time-dependently increased the systolic blood pressure (SBP) of mice, especially in DOCA-salt LID mice. Besides, the serum IGF-1 levels in WT mice were decreased after DOCA-salt induction. In addition, the plasma NE concentration and NE spillover, urinary protein, urea nitrogen, creatinine and RSNA were remarkably elevated with severe left ventricular dysfunction, but the creatinine clearance was reduced in DOCA-salt mice, and these similar changes were obvious in DOCA-salt mice with IGF-1 deletion. Moreover, the DOCA-salt mice had tubular ectasia, glomerular fibrosis, interstitial cell infiltration, and increased arterial wall thickness, and the DOCA-salt LID mice were more serious in those aspects. Deletion of IGF-1 may lead to enhanced RSNA in DOCA-salt hypertensive mice, thereby further aggravating left ventricular dysfunction and renal damage.
Collapse
Affiliation(s)
- Bing Xiao
- Department of Cardiology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China.
| | | | | | | | | | | |
Collapse
|
18
|
Zhang H, Liu Y, Yan L, Wang S, Zhang M, Ma C, Zheng X, Chen H, Zhu D. Long noncoding RNA Hoxaas3 contributes to hypoxia-induced pulmonary artery smooth muscle cell proliferation. Cardiovasc Res 2019; 115:647-657. [DOI: 10.1093/cvr/cvy250] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
Abstract
Aims
Long noncoding RNAs (lncRNAs) are involved in the regulation of vascular smooth muscle cells and cardiovascular pathology. However, the contribution of lncRNAs to pulmonary hypertension (PH) remains largely unknown. The over-proliferation of pulmonary artery smooth muscle cells (PASMCs) causes pulmonary arterial smooth muscle hypertrophy and stenosis of the pulmonary vascular lumen, resulting in PH. Here, we investigated the biological role of a novel lncRNA, Hoxa cluster antisense RNA 3 (Hoxaas3), in the regulation of cell proliferation in PH.
Methods and results
Hoxaas3 was up-regulated in the lung vasculature of hypoxic mice and in PASMCs under hypoxic conditions. Histone H3 Lysine 9 acetylation of Hoxaas3 promoted gene expression. Moreover, high expression of Hoxaas3 was associated with cell proliferation and modulated cell cycle distribution by up-regulating Homeobox a3 at the mRNA and protein levels.
Conclusion
This study defined the role and mechanism of action of Hoxaas3 in the regulation of cell proliferation in PH, which should facilitate the development of new therapeutic strategies for the treatment of this disease.
Collapse
Affiliation(s)
- Hongyue Zhang
- College of Pharmacy, Harbin Medical University, No.157, Baojian Road, Harbin, Heilongjiang, China
- Central Laboratory of Harbin Medical University-Daqing, No.38, Xinyang Road, Daqing, Heilongjiang, China
| | - Ying Liu
- College of Pharmacy, Harbin Medical University, No.157, Baojian Road, Harbin, Heilongjiang, China
- Central Laboratory of Harbin Medical University-Daqing, No.38, Xinyang Road, Daqing, Heilongjiang, China
| | - Lixin Yan
- College of Pharmacy, Harbin Medical University, No.157, Baojian Road, Harbin, Heilongjiang, China
- Central Laboratory of Harbin Medical University-Daqing, No.38, Xinyang Road, Daqing, Heilongjiang, China
| | - Siqi Wang
- College of Pharmacy, Harbin University of Commerce, No.138, Tongda Road, Harbin, Heilongjiang, China
| | - Min Zhang
- College of Pharmacy, Harbin Medical University, No.157, Baojian Road, Harbin, Heilongjiang, China
- Central Laboratory of Harbin Medical University-Daqing, No.38, Xinyang Road, Daqing, Heilongjiang, China
| | - Cui Ma
- College of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, No.38, Xinyang Road, Daqing, Heilongjiang, China
| | - Xiaodong Zheng
- Department of Pathophysiology, Harbin Medical University-Daqing, Daqing, No.38, Xinyang Road, Daqing, Heilongjiang, China
| | - He Chen
- Department of Obstetrics and gynecology, The Second affiliated Hospital of Harbin Medical University, No.246, Xuefu Road, Harbin, Heilongjiang, China
| | - Daling Zhu
- College of Pharmacy, Harbin Medical University, No.157, Baojian Road, Harbin, Heilongjiang, China
- Central Laboratory of Harbin Medical University-Daqing, No.38, Xinyang Road, Daqing, Heilongjiang, China
| |
Collapse
|
19
|
Zhong Y, Catheline D, Houeijeh A, Sharma D, Du L, Besengez C, Deruelle P, Legrand P, Storme L. Maternal omega-3 PUFA supplementation prevents hyperoxia-induced pulmonary hypertension in the offspring. Am J Physiol Lung Cell Mol Physiol 2018; 315:L116-L132. [PMID: 29597832 DOI: 10.1152/ajplung.00527.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pulmonary hypertension (PH) and right ventricular hypertrophy (RVH) affect 16-25% of premature infants with bronchopulmonary dysplasia (BPD), contributing significantly to perinatal morbidity and mortality. Omega-3 polyunsaturated fatty acids (PUFA ω-3) can improve vascular remodeling, angiogenesis, and inflammation under pathophysiological conditions. However, the effects of PUFA ω-3 supplementation in BPD-associated PH are unknown. The present study aimed to evaluate the effects of PUFA ω-3 on pulmonary vascular remodeling, angiogenesis, and inflammatory response in a hyperoxia-induced rat model of PH. From embryonic day 15, pregnant Sprague-Dawley rats were supplemented daily with PUFA ω-3, PUFA ω-6, or normal saline (0.2 ml/day). After birth, pups were pooled, assigned as 12 per litter, randomly assigned to either air or continuous oxygen exposure (fraction of inspired oxygen = 85%) for 20 days, and then euthanized for pulmonary hemodynamic and morphometric analysis. We found that PUFA ω-3 supplementation improved survival, decreased right ventricular systolic pressure and RVH caused by hyperoxia, and significantly improved alveolarization, vascular remodeling, and vascular density. PUFA ω-3 supplementation produced a higher level of total ω-3 in lung tissue and breast milk and was found to reverse the reduced levels of VEGFA, VEGF receptor 2, angiopoietin-1 (ANGPT1), endothelial TEK tyrosine kinase, endothelial nitric oxide synthase, and nitric oxide concentrations in lung tissue and the increased ANGPT2 levels in hyperoxia-exposed rats. The beneficial effects of PUFA ω-3 in improving lung injuries were also associated with an inhibition of leukocyte infiltration and reduced expression of the proinflammatory cytokines IL-1β, IL-6, and TNF-α. These data indicate that maternal PUFA ω-3 supplementation strategies could effectively protect against infant PH induced by hyperoxia.
Collapse
Affiliation(s)
- Ying Zhong
- Perinatal Environment and Health, UPRES EA 4489, Université de Lille, Centre Hospitalier Régional Universitaire de Lille , Lille , France.,Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine , Hangzhou , China
| | - Daniel Catheline
- Laboratoire de Biochimie et Nutrition Humaine, Institut National de la Recherche Agronomique USC 2012, Institut Supérieur des Sciences Agronomiques, Agroalimentaires, Horticoles et du Paysage, Rennes , France
| | - Ali Houeijeh
- Perinatal Environment and Health, UPRES EA 4489, Université de Lille, Centre Hospitalier Régional Universitaire de Lille , Lille , France.,Department of Neonatology, Centre Hospitalier Régional Universitaire de Lille , Lille , France
| | - Dyuti Sharma
- Perinatal Environment and Health, UPRES EA 4489, Université de Lille, Centre Hospitalier Régional Universitaire de Lille , Lille , France.,Department of Pediatric Surgery, Centre Hospitalier Régional Universitaire de Lille , Lille , France
| | - Lizhong Du
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine , Hangzhou , China
| | - Capucine Besengez
- Perinatal Environment and Health, UPRES EA 4489, Université de Lille, Centre Hospitalier Régional Universitaire de Lille , Lille , France
| | - Philippe Deruelle
- Perinatal Environment and Health, UPRES EA 4489, Université de Lille, Centre Hospitalier Régional Universitaire de Lille , Lille , France.,Department of Obstetrics and Gynecology, Centre Hospitalier Régional Universitaire de Lille , Lille , France
| | - Philippe Legrand
- Laboratoire de Biochimie et Nutrition Humaine, Institut National de la Recherche Agronomique USC 2012, Institut Supérieur des Sciences Agronomiques, Agroalimentaires, Horticoles et du Paysage, Rennes , France
| | - Laurent Storme
- Perinatal Environment and Health, UPRES EA 4489, Université de Lille, Centre Hospitalier Régional Universitaire de Lille , Lille , France.,Department of Neonatology, Centre Hospitalier Régional Universitaire de Lille , Lille , France
| |
Collapse
|
20
|
Abstract
Pulmonary hypertension is defined as a resting mean pulmonary artery pressure of 25 mm Hg or above. This review deals with pulmonary arterial hypertension (PAH), a type of pulmonary hypertension that primarily affects the pulmonary vasculature. In PAH, the pulmonary vasculature is dynamically obstructed by vasoconstriction, structurally obstructed by adverse vascular remodeling, and pathologically non-compliant as a result of vascular fibrosis and stiffening. Many cell types are abnormal in PAH, including vascular cells (endothelial cells, smooth muscle cells, and fibroblasts) and inflammatory cells. Progress has been made in identifying the causes of PAH and approving new drug therapies. A cancer-like increase in cell proliferation and resistance to apoptosis reflects acquired abnormalities of mitochondrial metabolism and dynamics. Mutations in the type II bone morphogenetic protein receptor (BMPR2) gene dramatically increase the risk of developing heritable PAH. Epigenetic dysregulation of DNA methylation, histone acetylation, and microRNAs also contributes to disease pathogenesis. Aberrant bone morphogenetic protein signaling and epigenetic dysregulation in PAH promote cell proliferation in part through induction of a Warburg mitochondrial-metabolic state of uncoupled glycolysis. Complex changes in cytokines (interleukins and tumor necrosis factor), cellular immunity (T lymphocytes, natural killer cells, macrophages), and autoantibodies suggest that PAH is, in part, an autoimmune, inflammatory disease. Obstructive pulmonary vascular remodeling in PAH increases right ventricular afterload causing right ventricular hypertrophy. In some patients, maladaptive changes in the right ventricle, including ischemia and fibrosis, reduce right ventricular function and cause right ventricular failure. Patients with PAH have dyspnea, reduced exercise capacity, exertional syncope, and premature death from right ventricular failure. PAH targeted therapies (prostaglandins, phosphodiesterase-5 inhibitors, endothelin receptor antagonists, and soluble guanylate cyclase stimulators), used alone or in combination, improve functional capacity and hemodynamics and reduce hospital admissions. However, these vasodilators do not target key features of PAH pathogenesis and have not been shown to reduce mortality, which remains about 50% at five years. This review summarizes the epidemiology, pathogenesis, diagnosis, and treatment of PAH.
Collapse
Affiliation(s)
| | - Mark L Ormiston
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - John J Ryan
- Department of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, ON, Canada
| |
Collapse
|
21
|
Loxham M, Smart DE, Bedke NJ, Smithers NP, Filippi I, Blume C, Swindle EJ, Tariq K, Howarth PH, Holgate ST, Davies DE. Allergenic proteases cleave the chemokine CX3CL1 directly from the surface of airway epithelium and augment the effect of rhinovirus. Mucosal Immunol 2018; 11:404-414. [PMID: 28677664 DOI: 10.1038/mi.2017.63] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 05/29/2017] [Indexed: 02/04/2023]
Abstract
CX3CL1 has been implicated in allergen-induced airway CD4+ T-lymphocyte recruitment in asthma. As epidemiological evidence supports a viral infection-allergen synergy in asthma exacerbations, we postulated that rhinovirus (RV) infection in the presence of allergen augments epithelial CX3CL1 release. Fully differentiated primary bronchial epithelial cultures were pretreated apically with house dust mite (HDM) extract and infected with rhinovirus-16 (RV16). CX3CL1 was measured by enzyme-linked immunosorbent assay and western blotting, and shedding mechanisms assessed using inhibitors, protease-activated receptor-2 (PAR-2) agonist, and recombinant CX3CL1-expressing HEK293T cells. Basolateral CX3CL1 release was unaffected by HDM but stimulated by RV16; inhibition by fluticasone or GM6001 implicated nuclear factor-κB and ADAM (A Disintegrin and Metalloproteinase) sheddases. Conversely, apical CX3CL1 shedding was stimulated by HDM and augmented by RV16. Although fluticasone or GM6001 reduced RV16+HDM-induced apical CX3CL1 release, heat inactivation or cysteine protease inhibition completely blocked CX3CL1 shedding. The HDM effect was via enzymatic cleavage of CX3CL1, not PAR-2 activation, yielding a product mitogenic for smooth muscle cells. Extracts of Alternaria fungus caused similar CX3CL1 shedding. We have identified a novel mechanism whereby allergenic proteases cleave CX3CL1 from the apical epithelial surface to yield a biologically active product. RV16 infection augmented HDM-induced CX3CL1 shedding-this may contribute to synergy between allergen exposure and RV infection in triggering asthma exacerbations and airway remodeling.
Collapse
Affiliation(s)
- M Loxham
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK.,Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton, UK
| | - D E Smart
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK
| | - N J Bedke
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK
| | - N P Smithers
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK
| | - I Filippi
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK.,Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - C Blume
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK
| | - E J Swindle
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK.,Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton, UK
| | - K Tariq
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK.,NIHR Southampton Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK
| | - P H Howarth
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK.,NIHR Southampton Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK
| | - S T Holgate
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK
| | - D E Davies
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK.,Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton, UK.,NIHR Southampton Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK
| |
Collapse
|
22
|
Bazan IS, Mensah KA, Rudkovskaia AA, Adonteng-Boateng PK, Herzog EL, Buckley L, Fares WH. Pulmonary arterial hypertension in the setting of scleroderma is different than in the setting of lupus: A review. Respir Med 2017; 134:42-46. [PMID: 29413506 DOI: 10.1016/j.rmed.2017.11.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 11/12/2017] [Accepted: 11/28/2017] [Indexed: 01/08/2023]
Abstract
Pulmonary hypertension (PH) is a clinical syndrome that is subdivided into five groups per the World Health Organization (WHO) classification, based largely on hemodynamic and pathophysiologic criteria. WHO Group 1 PH, termed pulmonary arterial hypertension (PAH), is a clinically progressive disease that can eventually lead to right heart failure and death, and it is hemodynamically characterized by pre-capillary PH and increased pulmonary vascular resistance in the absence of elevated left ventricular filling pressures. PAH can be idiopathic, heritable, or associated with a variety of conditions. Connective tissue diseases make up the largest portion of these associated conditions, most commonly systemic sclerosis (SSc), followed by mixed connective tissue disease and systemic lupus erythematous. These etiologies (namely SSc and Lupus) have been grouped together as connective tissue disease-associated PAH, however emerging evidence suggests they differ in pathogenesis, clinical course, prognosis, and treatment response. This review highlights the differences between SSc-PAH and Lupus-PAH. After introducing the diagnosis, screening, and pathobiology of PAH, we discuss connective tissue disease-associated PAH as a group, and then explore SSc-PAH and SLE-PAH separately, comparing these 2 PAH etiologies.
Collapse
Affiliation(s)
- Isabel S Bazan
- Yale University, School of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, New Haven, CT, USA
| | - Kofi A Mensah
- Yale University, School of Medicine, Section of Rheumatology, New Haven, CT, USA
| | | | | | - Erica L Herzog
- Yale University, School of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, New Haven, CT, USA
| | - Lenore Buckley
- Yale University, School of Medicine, Section of Rheumatology, New Haven, CT, USA
| | - Wassim H Fares
- Yale University, School of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, New Haven, CT, USA.
| |
Collapse
|
23
|
Jafri S, Ormiston ML. Immune regulation of systemic hypertension, pulmonary arterial hypertension, and preeclampsia: shared disease mechanisms and translational opportunities. Am J Physiol Regul Integr Comp Physiol 2017; 313:R693-R705. [PMID: 28978513 DOI: 10.1152/ajpregu.00259.2017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/11/2017] [Accepted: 10/02/2017] [Indexed: 12/22/2022]
Abstract
Systemic hypertension, preeclampsia, and pulmonary arterial hypertension (PAH) are diseases of high blood pressure in the systemic or pulmonary circulation. Beyond the well-defined contribution of more traditional pathophysiological mechanisms, such as changes in the renin-angiotensin-aldosterone system, to the development of these hypertensive disorders, there is substantial clinical evidence supporting an important role for inflammation and immunity in the pathogenesis of each of these three conditions. Over the last decade, work in small animal models, bearing targeted deficiencies in specific cytokines or immune cell subsets, has begun to clarify the immune-mediated mechanisms that drive changes in vascular structure and tone in hypertensive disease. By summarizing the clinical and experimental evidence supporting a contribution of the immune system to systemic hypertension, preeclampsia, and PAH, the current review highlights the cellular and molecular pathways that are common to all three hypertensive disorders. These mechanisms are centered on an imbalance in CD4+ helper T cell populations, defined by excessive Th17 responses and impaired Treg activity, as well as the excessive activation or impairment of additional immune cell types, including macrophages, dendritic cells, CD8+ T cells, B cells, and natural killer cells. The identification of common immune mechanisms in systemic hypertension, preeclampsia, and PAH raises the possibility of new therapeutic strategies that target the immune component of hypertension across multiple disorders.
Collapse
Affiliation(s)
- Salema Jafri
- University of Cambridge, Department of Medicine, Cambridge, United Kingdom; and
| | - Mark L Ormiston
- Queen's University, Departments of Biomedical and Molecular Sciences, Medicine and Surgery, Kingston, Canada
| |
Collapse
|
24
|
Middleton RC, Fournier M, Xu X, Marbán E, Lewis MI. Therapeutic benefits of intravenous cardiosphere-derived cell therapy in rats with pulmonary hypertension. PLoS One 2017; 12:e0183557. [PMID: 28837618 PMCID: PMC5570343 DOI: 10.1371/journal.pone.0183557] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/07/2017] [Indexed: 11/24/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive condition characterized by occlusive pulmonary arteriopathy, in which survival remains poor despite pharmacologic advances. The aim of this study was to evaluate the ability of cardiosphere-derived cells (CDCs), cardiac progenitor cells with potent anti-inflammatory and immunomodulatory properties, to attenuate hemodynamic and morphometric remodeling of the right ventricle (RV) and pulmonary arterioles in rats with established monocrotaline (MCT)-induced PAH. Animals were divided into 3 groups: 1) Control (CTL), 2) PAH in which CDCs were centrally infused (CDC) and 3) PAH in which saline was given (Sham). Significant increments in RV systolic pressure (RVSP) and RV hypertrophy were noted in Sham animals compared to CTL. In CDC rats at day 35, RSVP fell (- 38%; p< 0.001) and RV hypertrophy decreased (-26%; p< 0.01). TAPSE and cardiac output were preserved in all 3 groups at day 35. Pulmonary arteriolar wall thickness was greater in Sham rats compared to CTL, and reduced in CDC animals for vessels 20–50 μm (P<0.01; back to CTL levels) and 50–80μm (P<0.01) in diameter. The macrophage population was increased in Sham animals compared to CTL (P< 0.001), but markedly reduced in CDC rats. In conclusion, infusion of CDCs markedly attenuated several key pathophysiologic features of PAH. As adjunctive therapy to PAH-specific agents, CDCs have the potential to impact on the pathobiology of adverse pulmonary arteriolar remodeling, by acting on multiple mechanisms simultaneously.
Collapse
Affiliation(s)
- Ryan C. Middleton
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Mario Fournier
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
- Division of Pulmonary/Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Xuan Xu
- Division of Pulmonary/Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Eduardo Marbán
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Michael I. Lewis
- Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
- Division of Pulmonary/Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
- * E-mail:
| |
Collapse
|
25
|
Microarray Analysis and Detection of MicroRNAs Associated with Chronic Thromboembolic Pulmonary Hypertension. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8529796. [PMID: 28904974 PMCID: PMC5585581 DOI: 10.1155/2017/8529796] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/28/2017] [Accepted: 06/11/2017] [Indexed: 11/17/2022]
Abstract
The aim of this study was to understand the importance of chronic thromboembolic pulmonary hypertension- (CTEPH-) associated microRNAs (miRNAs). miRNAs differentially expressed in CTEPH samples compared with control samples were identified, and the target genes were predicted. The target genes of the key differentially expressed miRNAs were analyzed, and functional enrichment analyses were carried out. Finally, the miRNAs were detected using RT-PCR. Among the downregulated miRNAs, MiR-3148 regulated the most target genes and was significantly enriched in pathways in cancer, glioma, and ErbB signaling pathway. Furthermore, the number of target genes coregulated by miR-3148 and other miRNAs was the most. AR (androgen receptor), a target gene of hsa-miR-3148, was enriched in pathways in cancer. PRKCA (Protein Kinase C Alpha), also a target gene of hsa-miR-3148, was enriched in 15 of 16 KEGG pathways, such as pathways in cancer, glioma, and ErbB signaling pathway. In addition, the RT-PCR results showed that the expression of hsa-miR-3148 in CTEPH samples was significantly lower than that in control samples (P < 0.01). MiR-3148 may play an important role in the development of CTEPH. The key mechanisms for this miRNA may be hsa-miR-3148-AR-pathways in cancer or hsa-miR-3148-PRKCA-pathways in cancer/glioma/ErbB signaling pathway.
Collapse
|
26
|
Amsellem V, Abid S, Poupel L, Parpaleix A, Rodero M, Gary-Bobo G, Latiri M, Dubois-Rande JL, Lipskaia L, Combadiere C, Adnot S. Roles for the CX3CL1/CX3CR1 and CCL2/CCR2 Chemokine Systems in Hypoxic Pulmonary Hypertension. Am J Respir Cell Mol Biol 2017; 56:597-608. [PMID: 28125278 DOI: 10.1165/rcmb.2016-0201oc] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Monocytes/macrophages are major effectors of lung inflammation associated with various forms of pulmonary hypertension (PH). Interactions between the CCL2/CCR2 and CX3CL1/CX3CR1 chemokine systems that guide phagocyte infiltration are incompletely understood. Our objective was to explore the individual and combined actions of CCL2/CCR2 and CX3CL1/CX3CR1 in hypoxia-induced PH in mice; particularly their roles in monocyte trafficking, macrophage polarization, and pulmonary vascular remodeling. The development of hypoxia-induced PH was associated with marked increases in lung levels of CX3CR1, CCR2, and their respective ligands, CX3CL1 and CCL2. Flow cytometry revealed that both inflammatory Ly6Chi and resident Ly6Clo monocyte subsets exhibited sustained increases in blood and a transient peak in lung tissue, and that lung perivascular and alveolar macrophage counts showed sustained elevations. CX3CR1-/- mice were protected against hypoxic PH compared with wild-type mice, whereas CCL2-/- mice and double CX3CR1-/-/CCL2-/- mice exhibited similar PH severity, as did wild-type mice. The protective effects of CX3CR1 deficiency occurred concomitantly with increases in lung monocyte and macrophage counts and with a change from M2 to M1 macrophage polarization that markedly diminished the ability of conditioned media to induce pulmonary artery smooth muscle cell (PA-SMC) proliferation, which was partly dependent on CX3CL1 secretion. Results in mice given the CX3CR1 inhibitor F1 were similar to those in CX3CR1-/- mice. In conclusion, CX3CR1 deficiency protects against hypoxia-induced PH by modulating monocyte recruitment, macrophage polarization, and PA-SMC cell proliferation. Targeting CX3CR1 may hold promise for treating PH.
Collapse
Affiliation(s)
- Valérie Amsellem
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Shariq Abid
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Lucie Poupel
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Aurélien Parpaleix
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Mathieu Rodero
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Guillaume Gary-Bobo
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Mehdi Latiri
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Jean-Luc Dubois-Rande
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Larissa Lipskaia
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Christophe Combadiere
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Serge Adnot
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| |
Collapse
|
27
|
Miao R, Wang Y, Wan J, Leng D, Gong J, Li J, Liang Y, Zhai Z, Yang Y. Microarray expression profile of circular RNAs in chronic thromboembolic pulmonary hypertension. Medicine (Baltimore) 2017; 96:e7354. [PMID: 28682884 PMCID: PMC5502157 DOI: 10.1097/md.0000000000007354] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Chronic thromboembolic pulmonary hypertension (CTEPH) is a rare but debilitating and life-threatening complication of acute pulmonary embolism. Circular RNAs (circRNAs), presenting as covalently closed continuous loops, are RNA molecules with covalently joined 3'- and 5'-ends formed by back-splicing events. circRNAs may be significant biological molecules to understand disease mechanisms and to identify biomarkers for disease diagnosis and therapy. The aim of this study was to investigate the potential roles of circRNAs in CTEPH. METHODS Ten human blood samples (5 each from CTEPH and control groups) were included in the Agilent circRNA chip. The differentially expressed circRNAs were evaluated using t test, with significance set at a P value of < .05. A functional enrichment analysis for differentially expressed circRNAs was performed using DAVID online tools, and a Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis for target genes of miRNAs was performed using the R package clusterProfiler. Furthermore, miRNAs that interacted with differentially expressed circRNAs were predicted using the miRanda package. mRNAs that had clear biological functions and were regulated by miRNAs were predicted using miRWalk2.0 and then combined into a circRNA-miRNA-mRNA network. RESULTS In total, 351 differentially expressed circRNAs (122 upregulated and 229 downregulated) between CTEPH and control groups were obtained; among these circRNAs, hsa_circ_0002062 and hsa_circ_0022342 might be important because they can regulate 761 (e.g., hsa-miR-942-5p) and 453 (e.g., hsa-miR-940) miRNAs, respectively. Target genes (e.g., cyclin-dependent kinase 6) of hsa-miR-942-5p were mainly enriched in cancer-related pathways, whereas target genes (e.g., CRK-Like Proto-Oncogene, Adaptor Protein) of hsa-miR-940 were enriched in the ErbB signaling pathway. Therefore, these pathways are potentially important in CTEPH. CONCLUSIONS Our findings suggested that hsa_circ_0002062 and hsa_circ_0022342 may be key circRNAs for CTEPH development and that their targeted regulation may be an effective approach for treating CTEPH.
Collapse
Affiliation(s)
- Ran Miao
- Department of Clinical Laboratory, Beijing Chao-Yang Hospital, Capital Medical University
- Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Institute of Respiratory Medicine
| | - Ying Wang
- Department of Clinical Laboratory, Beijing Chao-Yang Hospital, Capital Medical University
- Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Institute of Respiratory Medicine
| | - Jun Wan
- Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Institute of Respiratory Medicine
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital
| | - Dong Leng
- Department of Clinical Laboratory, Beijing Chao-Yang Hospital, Capital Medical University
- Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Institute of Respiratory Medicine
| | - Juanni Gong
- Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Institute of Respiratory Medicine
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jifeng Li
- Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Institute of Respiratory Medicine
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yan Liang
- Department of Clinical Laboratory, Beijing Chao-Yang Hospital, Capital Medical University
- Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Institute of Respiratory Medicine
| | - Zhenguo Zhai
- Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Institute of Respiratory Medicine
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital
| | - Yuanhua Yang
- Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Institute of Respiratory Medicine
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Huhtinen A, Hongisto V, Laiho A, Löyttyniemi E, Pijnenburg D, Scheinin M. Gene expression profiles and signaling mechanisms in α 2B-adrenoceptor-evoked proliferation of vascular smooth muscle cells. BMC SYSTEMS BIOLOGY 2017; 11:65. [PMID: 28659168 PMCID: PMC5490158 DOI: 10.1186/s12918-017-0439-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 06/09/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND α2-adrenoceptors are important regulators of vascular tone and blood pressure. Regulation of cell proliferation is a less well investigated consequence of α2-adrenoceptor activation. We have previously shown that α2B-adrenoceptor activation stimulates proliferation of vascular smooth muscle cells (VSMCs). This may be important for blood vessel development and plasticity and for the pathology and therapeutics of cardiovascular disorders. The underlying cellular mechanisms have remained mostly unknown. This study explored pathways of regulation of gene expression and intracellular signaling related to α2B-adrenoceptor-evoked VSMC proliferation. RESULTS The cellular mechanisms and signaling pathways of α2B-adrenoceptor-evoked proliferation of VSMCs are complex and include redundancy. Functional enrichment analysis and pathway analysis identified differentially expressed genes associated with α2B-adrenoceptor-regulated VSMC proliferation. They included the upregulated genes Egr1, F3, Ptgs2 and Serpine1 and the downregulated genes Cx3cl1, Cav1, Rhoa, Nppb and Prrx1. The most highly upregulated gene, Lypd8, represents a novel finding in the VSMC context. Inhibitor library screening and kinase activity profiling were applied to identify kinases in the involved signaling pathways. Putative upstream kinases identified by two different screens included PKC, Raf-1, Src, the MAP kinases p38 and JNK and the receptor tyrosine kinases EGFR and HGF/HGFR. As a novel finding, the Src family kinase Lyn was also identified as a putative upstream kinase. CONCLUSIONS α2B-adrenoceptors may mediate their pro-proliferative effects in VSMCs by promoting the activity of bFGF and PDGF and the growth factor receptors EGFR, HGFR and VEGFR-1/2. The Src family kinase Lyn was also identified as a putative upstream kinase. Lyn is known to be expressed in VSMCs and has been identified as an important regulator of GPCR trafficking and GPCR effects on cell proliferation. Identified Ser/Thr kinases included several PKC isoforms and the β-adrenoceptor kinases 1 and 2. Cross-talk between the signaling mechanisms involved in α2B-adrenoceptor-evoked VSMC proliferation thus appears to involve PKC activation, subsequent changes in gene expression, transactivation of EGFR, and modulation of kinase activities and growth factor-mediated signaling. While many of the identified individual signals were relatively small in terms of effect size, many of them were validated by combining pathway analysis and our integrated screening approach.
Collapse
Affiliation(s)
- Anna Huhtinen
- Department of Pharmacology, Drug Development and Therapeutics, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Vesa Hongisto
- Toxicology Division, Misvik Biology Oy, Turku, Finland
| | - Asta Laiho
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Eliisa Löyttyniemi
- Department of Biostatistics, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Dirk Pijnenburg
- PamGene International BV, Wolvenhoek 10, 5211HH s’Hertogenbosch, The Netherlands
| | - Mika Scheinin
- Department of Pharmacology, Drug Development and Therapeutics, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| |
Collapse
|
29
|
Sankhe S, Manousakidi S, Antigny F, Arthur Ataam J, Bentebbal S, Ruchon Y, Lecerf F, Sabourin J, Price L, Fadel E, Dorfmüller P, Eddahibi S, Humbert M, Perros F, Capuano V. T-type Ca 2+ channels elicit pro-proliferative and anti-apoptotic responses through impaired PP2A/Akt1 signaling in PASMCs from patients with pulmonary arterial hypertension. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1631-1641. [PMID: 28655554 DOI: 10.1016/j.bbamcr.2017.06.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/13/2017] [Accepted: 06/21/2017] [Indexed: 10/19/2022]
Abstract
Idiopathic pulmonary arterial hypertension (iPAH) is characterized by obstructive hyperproliferation and apoptosis resistance of distal pulmonary artery smooth muscle cells (PASMCs). T-type Ca2+ channel blockers have been shown to reduce experimental pulmonary hypertension, although the impact of T-type channel inhibition remains unexplored in PASMCs from iPAH patients. Here we show that T-type channels Cav3.1 and Cav3.2 are present in the lung and PASMCs from iPAH patients and control subjects. The blockade of T-type channels by the specific blocker, TTA-A2, prevents cell cycle progression and PASMCs growth. In iPAH cells, T-type channel signaling fails to activate phosphatase PP2A, leading to an increase in ERK1/2, P38 activation. Moreover, T-type channel signaling is redirected towards the activation of the kinase Akt1, leading to increased expression of the anti-apoptotic protein survivin, and a decrease in the pro-apoptotic mediator FoxO3A. Finally, in iPAH cells, Akt1 is no longer able to regulate caspase 9 activation, whereas T-type channel overexpression reverses PP2A defect in iPAH cells but reinforces the deleterious effects of Akt1 activation. Altogether, these data highlight T-type channel signaling as a strong trigger of the pathological phenotype of PASMCs from iPAH patients (hyper-proliferation/cells survival and apoptosis resistance), suggesting that both T-type channels and PP2A may be promising therapeutic targets for pulmonary hypertension.
Collapse
Affiliation(s)
- Safietou Sankhe
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Sevasti Manousakidi
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Fabrice Antigny
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Jennifer Arthur Ataam
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Sana Bentebbal
- PhyMedExp, Univ. Montpellier, Inserm U1046, cNRS UMR9214.34295 MINSERM U1046, Montpellier, France
| | - Yann Ruchon
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Florence Lecerf
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Jessica Sabourin
- INSERM UMR-S1180, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Laura Price
- National Pulmonary Hypertension Service, Royal Brompton Hospital, Sydney Street, London SW3 6NP, UK
| | - Elie Fadel
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Peter Dorfmüller
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Saadia Eddahibi
- PhyMedExp, Univ. Montpellier, Inserm U1046, cNRS UMR9214.34295 MINSERM U1046, Montpellier, France
| | - Marc Humbert
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France; AP-HP, Service de pneumologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Frédéric Perros
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Véronique Capuano
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France.
| |
Collapse
|
30
|
Abstract
OBJECTIVE The present study aims to identify the role of inflammatory markers such as C-reactive protein, interleukin-6, and fractalkine in CHD-associated pulmonary hypertension in children. METHODS This is a prospective review of 37 children with CHD-related pulmonary hypertension, 21 children with congenital heart defects, and 22 healthy children. RESULTS Serum C-reactive protein and interleukin-6 levels were significantly higher in the children with CHD-related pulmonary hypertension (respectively, p=0.049 and 0.026). Serum C-reactive protein concentrations correlated negatively with ejection fraction (r=-0.609, p=0.001) and fractional shortening (r=-0.452, p=0.007) in the pulmonary hypertension group. Serum fractalkine concentrations correlated negatively with ejection fraction (r=-0.522, p=0.002) and fractional shortening (r=-0.395, p=0.021) in the children with pulmonary hypertension. Serum interleukin-6 concentrations also correlated negatively with Qs (r=-0.572, p=0.021), positively with Rs (r=0.774, p=0.001), and positively with pulmonary wedge pressure (r=0.796, p=0.006) in the pulmonary hypertension group. A cut-off value of 2.2 IU/L for C-reactive protein was able to predict pulmonary hypertension with 77.5% sensitivity and 77.5% specificity. When the cut-off point for interleukin-6 concentration was 57.5 pg/ml, pulmonary hypertension could be predicted with 80% sensitivity and 75% specificity. CONCLUSION Inflammation is associated with the pathophysiology of pulmonary hypertension. The inflammatory markers C-reactive protein and interleukin-6 may have a role in the clinical evaluation of paediatric pulmonary hypertension related to CHDs.
Collapse
|
31
|
Di Mise A, Wang YX, Zheng YM. Role of Transcription Factors in Pulmonary Artery Smooth Muscle Cells: An Important Link to Hypoxic Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:13-32. [PMID: 29047078 DOI: 10.1007/978-3-319-63245-2_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hypoxia, namely a lack of oxygen in the blood, induces pulmonary vasoconstriction and vasoremodeling, which serve as essential pathologic factors leading to pulmonary hypertension (PH). The underlying molecular mechanisms are uncertain; however, pulmonary artery smooth muscle cells (PASMCs) play an essential role in hypoxia-induced pulmonary vasoconstriction, vasoremodeling, and PH. Hypoxia causes oxidative damage to DNAs, proteins, and lipids. This damage (oxidative stress) modulates the activity of ion channels and elevates the intracellular calcium concentration ([Ca2+]i, Ca2+ signaling) of PASMCs. The oxidative stress and increased Ca2+ signaling mutually interact with each other, and synergistically results in a variety of cellular responses. These responses include functional and structural abnormalities of mitochondria, sarcoplasmic reticulum, and nucleus; cell contraction, proliferation, migration, and apoptosis, as well as generation of vasoactive substances, inflammatory molecules, and growth factors that mediate the development of PH. A number of studies reveal that various transcription factors (TFs) play important roles in hypoxia-induced oxidative stress, disrupted PAMSC Ca2+ signaling and the development and progress of PH. It is believed that in the pathogenesis of PH, hypoxia facilitates these roles by mediating the expression of multiple genes. Therefore, the identification of specific genes and their transcription factors implicated in PH is necessary for the complete understanding of the underlying molecular mechanisms. Moreover, this identification may aid in the development of novel and effective therapeutic strategies for PH.
Collapse
Affiliation(s)
- Annarita Di Mise
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Yong-Xiao Wang
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| | - Yun-Min Zheng
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| |
Collapse
|
32
|
Zhang X, Hou HT, Wang J, Liu XC, Yang Q, He GW. Plasma Proteomic Study in Pulmonary Arterial Hypertension Associated with Congenital Heart Diseases. Sci Rep 2016; 6:36541. [PMID: 27886187 PMCID: PMC5122864 DOI: 10.1038/srep36541] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 10/17/2016] [Indexed: 02/06/2023] Open
Abstract
Pulmonary arterial hypertension associated with congenital heart disease (CHD-PAH) has serious consequence and plasma protein profiles in CHD-PAH are unknown. We aimed to reveal the differential plasma proteins in 272 CHD patients with or without PAH. Various types of CHD-PAH were studied. Differential plasma proteins were first detected by iTRAQ proteomic technology and those with significant clinical relevance were selected for further ELISA validation in new cohort of patients. Among the 190 differential plasma proteins detected by iTRAQ, carbamoyl-phosphate synthetase I (CPSI, related to urea cycle and endogenous nitric oxide production) and complement factor H-related protein 2 (CFHR2, related to complement system and coagulant mechanism) were selected for further ELISA validation in new cohort of 152 patients. Both CPSI and CFHR2 were down-regulated with decreased plasma levels (p < 0.01). Thus, we for the first time in CHD-PAH patients identified a large number of differential plasma proteins. The decreased CPSI expression in CHD-PAH patients may reveal a mechanism related to endogenous nitric oxide and the decrease of CFHR2 protein may demonstrate the deficiency of the immune system and coagulation mechanism. The findings may open a new direction for translational medicine in CHD-PAH with regard to the diagnosis and progress of the disease.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Tianjin & The Affiliated Hospital of Hangzhou Normal University & Zhejiang University, Hangzhou, China
| | - Hai-Tao Hou
- TEDA International Cardiovascular Hospital, CAMS & PUMC, Tianjin, China
| | - Jun Wang
- TEDA International Cardiovascular Hospital, CAMS & PUMC, Tianjin, China
| | - Xiao-Cheng Liu
- TEDA International Cardiovascular Hospital, CAMS & PUMC, Tianjin, China
| | - Qin Yang
- TEDA International Cardiovascular Hospital, CAMS & PUMC, Tianjin, China
- The Chinese University of Hong Kong, Shatin, China
| | - Guo-Wei He
- Department of Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Tianjin & The Affiliated Hospital of Hangzhou Normal University & Zhejiang University, Hangzhou, China
- Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
33
|
Chen IC, Dai ZK. Insight into Pulmonary Arterial Hypertension Associated with Congenital Heart Disease (PAH-CHD): Classification and Pharmacological Management from a Pediatric Cardiological Point of View. ACTA CARDIOLOGICA SINICA 2016; 31:507-15. [PMID: 27122915 DOI: 10.6515/acs20150424b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
UNLABELLED Compared with adult patients with pulmonary hypertension (PH), pulmonary vascular disease is characterized by complex heterogeneity in pediatric patients. The Nice PH classification does not completely characterize or individualize any subgroup of pediatric PH. This is in contrast to the Panama classification, in which prenatal and fetal origins of many pulmonary vascular diseases in neonates and children, perinatal pulmonary vascular maladaptation, prenatal and postnatal pulmonary vascular mal-development, and pulmonary vascular hypoplasia are included. Currently, the updated treatment algorithm for adults with pulmonary arterial hypertension (PAH), including PAH associated with congenital heart disease (PAH-CHD) and idiopathic PAH, etc. has been reported. It has been suggested to treat FC III patients with Eisenmenger syndrome (ES) with bosentan. However, there is no evidence-based treatment algorithm for children with PAH-CHD. Moreover, it is necessary to develop a more comprehensive algorithm in which multiple specific pediatric risk factors are determined, and the critical goal of treatment should be to permit normal activities without the need to self-limit in children with PAH-CHD. Together, the beneficial data on specific-target pharmacologic interventions are still quite preliminary, and large trials are warranted. Specifically, the extrapolation of the other forms of the disease, such as ES, should be undertaken carefully. KEY WORDS Congenital heart disease; Eisenmenger syndrome; Pulmonary arterial hypertension; Target therapy.
Collapse
Affiliation(s)
- I-Chen Chen
- Department of Pediatrics, Division of Pediatric Cardiology and Pulmonology, Kaohsiung Medical University Hospital, Kaohsiung Medical University
| | - Zen-Kong Dai
- Department of Pediatrics, Division of Pediatric Cardiology and Pulmonology, Kaohsiung Medical University Hospital, Kaohsiung Medical University; ; Department of Pediatrics, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
34
|
Johnson SM, McNally BA, Ioannidis I, Flano E, Teng MN, Oomens AG, Walsh EE, Peeples ME. Respiratory Syncytial Virus Uses CX3CR1 as a Receptor on Primary Human Airway Epithelial Cultures. PLoS Pathog 2015; 11:e1005318. [PMID: 26658574 PMCID: PMC4676609 DOI: 10.1371/journal.ppat.1005318] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 11/10/2015] [Indexed: 01/16/2023] Open
Abstract
Respiratory syncytial virus (RSV) is the most frequent cause of lower respiratory disease in infants, but no vaccine or effective therapy is available. The initiation of RSV infection of immortalized cells is largely dependent on cell surface heparan sulfate (HS), a receptor for the RSV attachment (G) glycoprotein in immortalized cells. However, RSV infects the ciliated cells in primary well differentiated human airway epithelial (HAE) cultures via the apical surface, but HS is not detectable on this surface. Here we show that soluble HS inhibits infection of immortalized cells, but not HAE cultures, confirming that HS is not the receptor on HAE cultures. Conversely, a “non-neutralizing” monoclonal antibody against the G protein that does not block RSV infection of immortalized cells, does inhibit infection of HAE cultures. This antibody was previously shown to block the interaction between the G protein and the chemokine receptor CX3CR1 and we have mapped the binding site for this antibody to the CX3C motif and its surrounding region in the G protein. We show that CX3CR1 is present on the apical surface of ciliated cells in HAE cultures and especially on the cilia. RSV infection of HAE cultures is reduced by an antibody against CX3CR1 and by mutations in the G protein CX3C motif. Additionally, mice lacking CX3CR1 are less susceptible to RSV infection. These findings demonstrate that RSV uses CX3CR1 as a cellular receptor on HAE cultures and highlight the importance of using a physiologically relevant model to study virus entry and antibody neutralization. Respiratory syncytial virus (RSV) is the second most common infectious cause of infant death worldwide. Despite this great clinical impact, no effective antivirals or vaccines against RSV are available. Here we find that the RSV attachment (G) glycoprotein uses CX3CR1 as a receptor on primary human airway epithelial (HAE) cultures, an excellent model of RSV infection of the human lung. The G protein contains a CX3C motif and we find that this region is critical for its role in infection of HAE cultures, but not of immortalized cells. Furthermore, we find that antibodies against the G protein neutralize RSV infection of HAE cultures differently from immortalized cells. These insights suggest that HAE cultures should be used to quantify neutralizing antibodies, including during vaccine development, that the CX3CR1 interaction with the RSV G protein could be a target for antiviral drug development, and that the G protein should be considered for inclusion in vaccines.
Collapse
Affiliation(s)
- Sara M. Johnson
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Beth A. McNally
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Ioannis Ioannidis
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Emilio Flano
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Michael N. Teng
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Antonius G. Oomens
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Edward E. Walsh
- School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Mark E. Peeples
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
35
|
Dexamethasone induces apoptosis in pulmonary arterial smooth muscle cells. Respir Res 2015; 16:114. [PMID: 26382031 PMCID: PMC4574531 DOI: 10.1186/s12931-015-0262-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 08/18/2015] [Indexed: 01/30/2023] Open
Abstract
Background Dexamethasone suppressed inflammation and haemodynamic changes in an animal model of pulmonary arterial hypertension (PAH). A major target for dexamethasone actions is NF-κB, which is activated in pulmonary vascular cells and perivascular inflammatory cells in PAH. Reverse remodelling is an important concept in PAH disease therapy, and further to its anti-proliferative effects, we sought to explore whether dexamethasone augments pulmonary arterial smooth muscle cell (PASMC) apoptosis. Methods Analysis of apoptosis markers (caspase 3, in-situ DNA fragmentation) and NF-κB (p65 and phospho-IKK-α/β) activation was performed on lung tissue from rats with monocrotaline (MCT)-induced pulmonary hypertension (PH), before and after day 14–28 treatment with dexamethasone (5 mg/kg/day). PASMC were cultured from this rat PH model and from normal human lung following lung cancer surgery. Following stimulation with TNF-α (10 ng/ml), the effects of dexamethasone (10−8–10−6 M) and IKK2 (NF-κB) inhibition (AS602868, 0–3 μM (0-3×10−6 M) on IL-6 and CXCL8 release and apoptosis was determined by ELISA and by Hoechst staining. NF-κB activation was measured by TransAm assay. Results Dexamethasone treatment of rats with MCT-induced PH in vivo led to PASMC apoptosis as displayed by increased caspase 3 expression and DNA fragmentation. A similar effect was seen in vitro using TNF-α-simulated human and rat PASMC following both dexamethasone and IKK2 inhibition. Increased apoptosis was associated with a reduction in NF-κB activation and in IL-6 and CXCL8 release from PASMC. Conclusions Dexamethasone exerted reverse-remodelling effects by augmenting apoptosis and reversing inflammation in PASMC possibly via inhibition of NF-κB. Future PAH therapies may involve targeting these important inflammatory pathways.
Collapse
|
36
|
Li L, Wang X, Wang L, Qu L, Zhu X, Li M, Dang X, Li P, Gao Y, Peng Z, Pan L, Wan L. Mammalian target of rapamycin overexpression antagonizes chronic hypoxia-triggered pulmonary arterial hypertension via the autophagic pathway. Int J Mol Med 2015; 36:316-322. [PMID: 26017061 DOI: 10.3892/ijmm.2015.2224] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 05/18/2015] [Indexed: 02/05/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive pulmonary vascular disorder with high morbidity and mortality, and is characterized by excessive growth of endothelial cells. Recently, the mammalian target of rapamycin (mTOR) has attracted increasing attention due to its potential as a therapeutic target against certain diseases associated with proliferative and metabolic abnormalities. However, the effect on mTOR on PAH has not yet been elucidated. In the present study, a marked downregulation of mTOR was observed in PAH patients. Following construction of a mouse model of PAH by chronic exposure to hypoxia, adenovirus-mediated upregulation of mTOR significantly attenuated right ventricular systolic pressure, right ventricular hypertrophy and wall thickness of pulmonary arterioles, indicating a protective effect of mTOR on PAH. Further analysis confirmed that mTOR overexpression inhibited autophagy triggered by hypoxia through blocking light chain 3 II expression and increasing p62 levels. In vitro, hypoxia enhanced the proliferation of human pulmonary artery endothelial cells (PAECs), which was markedly abrogated by mTOR overexpression. Of note, upregulation of mTOR inhibited the hypoxia-induced autophagy pathway, which contributed to cell proliferation, while silencing of autophagy by RNA interference with ATG5 significantly inhibited cell proliferation. In conclusion, the results of the present study suggested a potential protective effect of mTOR on the progression of PAH by suppressing PAEC proliferation through blocking the autophagic pathway. Therefore, the present study suggested that mTOR is a promising therapeutic agent against PAH.
Collapse
Affiliation(s)
- Lingxia Li
- The Cadre Ward, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xiaochuang Wang
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Lina Wang
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Li Qu
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xinye Zhu
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Manxiang Li
- Department of Respiratory Diseases, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xiaoyan Dang
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Ping Li
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yanxia Gao
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Zhuo Peng
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Longfei Pan
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Li Wan
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
37
|
Chirkova T, Lin S, Oomens AGP, Gaston KA, Boyoglu-Barnum S, Meng J, Stobart CC, Cotton CU, Hartert TV, Moore ML, Ziady AG, Anderson LJ. CX3CR1 is an important surface molecule for respiratory syncytial virus infection in human airway epithelial cells. J Gen Virol 2015; 96:2543-2556. [PMID: 26297201 DOI: 10.1099/vir.0.000218] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of severe pneumonia and bronchiolitis in infants and young children, and causes disease throughout life. Understanding the biology of infection, including virus binding to the cell surface, should help develop antiviral drugs or vaccines. The RSV F and G glycoproteins bind cell surface heparin sulfate proteoglycans (HSPGs) through heparin-binding domains. The G protein also has a CX3C chemokine motif which binds to the fractalkine receptor CX3CR1. G protein binding to CX3CR1 is not important for infection of immortalized cell lines, but reportedly is so for primary human airway epithelial cells (HAECs), the primary site for human infection. We studied the role of CX3CR1 in RSV infection with CX3CR1-transfected cell lines and HAECs with variable percentages of CX3CR1-expressing cells, and the effect of anti-CX3CR1 antibodies or a mutation in the RSV CX3C motif. Immortalized cells lacking HSPGs had low RSV binding and infection, which was increased markedly by CX3CR1 transfection. CX3CR1 was expressed primarily on ciliated cells, and ∼50 % of RSV-infected cells in HAECs were CX3CR1+. HAECs with more CX3CR1-expressing cells had a proportional increase in RSV infection. Blocking G binding to CX3CR1 with anti-CX3CR1 antibody or a mutation in the CX3C motif significantly decreased RSV infection in HAECs. The kinetics of cytokine production suggested that the RSV/CX3CR1 interaction induced RANTES (regulated on activation normal T-cell expressed and secreted protein), IL-8 and fractalkine production, whilst it downregulated IL-15, IL1-RA and monocyte chemotactic protein-1. Thus, the RSV G protein/CX3CR1 interaction is likely important in infection and infection-induced responses of the airway epithelium, the primary site of human infection.
Collapse
Affiliation(s)
- Tatiana Chirkova
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Songbai Lin
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Antonius G P Oomens
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Kelsey A Gaston
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Seyhan Boyoglu-Barnum
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Jia Meng
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Christopher C Stobart
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Calvin U Cotton
- Division of Pediatric Pulmonology, Case Western University, Cleveland, Ohio, USA
| | - Tina V Hartert
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine and Vanderbilt Center for Asthma and Environmental Health Sciences Research, Vanderbilt University, Nashville, Tennessee, USA
| | - Martin L Moore
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Assem G Ziady
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Larry J Anderson
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
38
|
Increase of serum fractalkine and fractalkine gene expression levels in sickle cell disease patients. Int J Hematol 2014; 101:114-8. [DOI: 10.1007/s12185-014-1718-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/20/2014] [Accepted: 11/27/2014] [Indexed: 11/27/2022]
|
39
|
Vaillancourt M, Ruffenach G, Meloche J, Bonnet S. Adaptation and remodelling of the pulmonary circulation in pulmonary hypertension. Can J Cardiol 2014; 31:407-15. [PMID: 25630876 DOI: 10.1016/j.cjca.2014.10.023] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 10/06/2014] [Accepted: 10/20/2014] [Indexed: 01/22/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by remodelling of pulmonary arteries caused by a proliferation/apoptosis imbalance within the vascular wall. This pathological phenotype seems to be triggered by different environmental stress and injury events such as increased inflammation, DNA damage, and epigenetic deregulation. It appears that one of the first hit to occur is endothelial cells (ECs) injury and apoptosis, which leads to paracrine signalling to other ECs, pulmonary artery smooth muscle cells (PASMCs), and fibroblasts. These signals promote a phenotypic change of surviving ECs by disturbing different signalling pathways leading to sustained vasoconstriction, proproliferative and antiapoptotic phenotype, deregulated angiogenesis, and formation of plexiform lesions. EC signalling also recruits proinflammatory cells, leading to pulmonary infiltration of lymphocytes, macrophages, and dendritic cells, sustaining the inflammatory environment and autoimmune response. Finally, EC signalling promotes proliferative and antiapoptotic PAH-PASMC phenotypes, which acquire migratory capacities, resulting in increased vascular wall thickness and muscularization of small pulmonary arterioles. Adaptation and remodelling of pulmonary circulation also involves epigenetic components, such as microRNA deregulation, DNA methylation, and histone modification. This review will focus on the different cellular and epigenetic aspects including EC stress response, molecular mechanisms contributing to PAH-PASMC and PAEC proliferation and resistance to apoptosis, as well as epigenetic control involved in adaptation and remodelling of the pulmonary circulation in PAH.
Collapse
Affiliation(s)
- Mylène Vaillancourt
- Pulmonary Hypertension Research Group of The Quebec Heart And Lung Institute Research Centre, Québec City, Québec, Canada
| | - Grégoire Ruffenach
- Pulmonary Hypertension Research Group of The Quebec Heart And Lung Institute Research Centre, Québec City, Québec, Canada
| | - Jolyane Meloche
- Pulmonary Hypertension Research Group of The Quebec Heart And Lung Institute Research Centre, Québec City, Québec, Canada.
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group of The Quebec Heart And Lung Institute Research Centre, Québec City, Québec, Canada.
| |
Collapse
|
40
|
Zheng L, Cao Y, Liu S, Peng Z, Zhang S. Neferine inhibits angiotensin II-induced rat aortic smooth muscle cell proliferation predominantly by downregulating fractalkine gene expression. Exp Ther Med 2014; 8:1545-1550. [PMID: 25289057 PMCID: PMC4186444 DOI: 10.3892/etm.2014.1952] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 06/19/2014] [Indexed: 02/05/2023] Open
Abstract
Neferine inhibits the angiotensin II (AngII)-induced proliferation of vascular smooth muscle cells (SMCs), but the underlying mechanism is unclear. The aim of this study was to explore the mechanism underlying the effect of neferine on the proliferation of vascular SMCs. Rat aortic SMCs (RASMCs) were used and fractalkine (Fkn) gene expression was measured by quantitative polymerase chain reaction and western blot analysis. The proliferation of RASMCs was analyzed by MTT assay and flow cytometry. It was revealed that AngII induced Fkn expression in a dose- and time-dependent manner. Fkn-knockdown with small interfering RNA attenuated the AngII-induced RASMC proliferation. Furthermore, neferine inhibited Fkn expression and attenuated the AngII-induced RASMC proliferation. These findings suggest that the Fkn gene may play an important role in AngII-induced RASMC proliferation and that neferine acts to attenuate AngII-induced RASMC proliferation by inhibiting Fkn expression.
Collapse
Affiliation(s)
- Lulu Zheng
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yongwen Cao
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Shao Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Zhenyu Peng
- Department of Emergency, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Saidan Zhang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
41
|
Rabinovitch M, Guignabert C, Humbert M, Nicolls MR. Inflammation and immunity in the pathogenesis of pulmonary arterial hypertension. Circ Res 2014; 115:165-75. [PMID: 24951765 DOI: 10.1161/circresaha.113.301141] [Citation(s) in RCA: 741] [Impact Index Per Article: 67.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This review summarizes an expanding body of knowledge indicating that failure to resolve inflammation and altered immune processes underlie the development of pulmonary arterial hypertension. The chemokines and cytokines implicated in pulmonary arterial hypertension that could form a biomarker platform are discussed. Pre-clinical studies that provide the basis for dysregulated immunity in animal models of the disease are reviewed. In addition, we present therapies that target inflammatory/immune mechanisms that are currently enrolling patients, and discuss others in development. We show how genetic and metabolic abnormalities are inextricably linked to dysregulated immunity and adverse remodeling in the pulmonary arteries.
Collapse
Affiliation(s)
- Marlene Rabinovitch
- From the Cardiovascular Institute and Department of Pediatrics (M.R.) and Department of Medicine (M.R.N.), Stanford University School of Medicine, CA; INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson and Université Paris-Sud, School of Medicine, Le Kremlin-Bicêtre (C.G., M.H.); and AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, France (M.H.).
| | - Christophe Guignabert
- From the Cardiovascular Institute and Department of Pediatrics (M.R.) and Department of Medicine (M.R.N.), Stanford University School of Medicine, CA; INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson and Université Paris-Sud, School of Medicine, Le Kremlin-Bicêtre (C.G., M.H.); and AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, France (M.H.)
| | - Marc Humbert
- From the Cardiovascular Institute and Department of Pediatrics (M.R.) and Department of Medicine (M.R.N.), Stanford University School of Medicine, CA; INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson and Université Paris-Sud, School of Medicine, Le Kremlin-Bicêtre (C.G., M.H.); and AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, France (M.H.)
| | - Mark R Nicolls
- From the Cardiovascular Institute and Department of Pediatrics (M.R.) and Department of Medicine (M.R.N.), Stanford University School of Medicine, CA; INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson and Université Paris-Sud, School of Medicine, Le Kremlin-Bicêtre (C.G., M.H.); and AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, France (M.H.)
| |
Collapse
|
42
|
Shao D, Perros F, Caramori G, Meng C, Dormuller P, Chou PC, Church C, Papi A, Casolari P, Welsh D, Peacock A, Humbert M, Adcock IM, Wort SJ. Nuclear IL-33 regulates soluble ST2 receptor and IL-6 expression in primary human arterial endothelial cells and is decreased in idiopathic pulmonary arterial hypertension. Biochem Biophys Res Commun 2014; 451:8-14. [DOI: 10.1016/j.bbrc.2014.06.111] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 06/23/2014] [Indexed: 11/26/2022]
|
43
|
Amsellem V, Lipskaia L, Abid S, Poupel L, Houssaini A, Quarck R, Marcos E, Mouraret N, Parpaleix A, Bobe R, Gary-Bobo G, Saker M, Dubois-Randé JL, Gladwin MT, Norris KA, Delcroix M, Combadière C, Adnot S. CCR5 as a treatment target in pulmonary arterial hypertension. Circulation 2014; 130:880-891. [PMID: 24993099 DOI: 10.1161/circulationaha.114.010757] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Pulmonary arterial hypertension (PH), whether idiopathic or related to underlying diseases such as HIV infection, results from complex vessel remodeling involving both pulmonary artery smooth muscle cell (PA-SMC) proliferation and inflammation. CCR5, a coreceptor for cellular HIV-1 entry expressed on macrophages and vascular cells, may be involved in the pathogenesis of PH. Maraviroc is a new CCR5 antagonist designed to block HIV entry. METHODS AND RESULTS Marked CCR5 expression was found in lungs from patients with idiopathic PH, in mice with hypoxia-induced PH, and in Simian immunodeficiency virus-infected macaques, in which it was localized chiefly in the PA-SMCs. To assess the role for CCR5 in experimental PH, we used both gene disruption and pharmacological CCR5 inactivation in mice. Because maraviroc does not bind to murine CCR5, we used human-CCR5ki mice for pharmacological and immunohistochemical studies. Compared with wild-type mice, CCR5-/- mice or human-CCR5ki mice treated with maraviroc exhibited decreased PA-SMC proliferation and recruitment of perivascular and alveolar macrophages during hypoxia exposure. CCR5-/- mice reconstituted with wild-type bone marrow cells and wild-type mice reconstituted with CCR5-/- bone marrow cells were protected against PH, suggesting CCR5-mediated effects on PA-SMCs and macrophage involvement. The CCR5 ligands CCL5 and the HIV-1 gp120 protein increased intracellular calcium and induced growth of human and human-CCR5ki mouse PA-SMCs; maraviroc inhibited both effects. Maraviroc also reduced the growth-promoting effects of conditioned media from CCL5-activated macrophages derived from human-CCR5ki mice on PA-SMCs from wild-type mice. CONCLUSION The CCL5-CCR5 pathway represents a new therapeutic target in PH associated with HIV or with other conditions.
Collapse
Affiliation(s)
- Valérie Amsellem
- Inserm U955 and Département de Physiologie, Hôpital Henri Mondor, Créteil, France, Université Paris-Est Créteil (UPEC), France
| | - Larissa Lipskaia
- Inserm U955 and Département de Physiologie, Hôpital Henri Mondor, Créteil, France, Université Paris-Est Créteil (UPEC), France
| | - Shariq Abid
- Inserm U955 and Département de Physiologie, Hôpital Henri Mondor, Créteil, France, Université Paris-Est Créteil (UPEC), France
| | - Lucie Poupel
- Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Amal Houssaini
- Inserm U955 and Département de Physiologie, Hôpital Henri Mondor, Créteil, France, Université Paris-Est Créteil (UPEC), France
| | - Rozenn Quarck
- Respiratory Division, University Hospitals of Leuven and Department of Clinical and Experimental Medicine, University of Leuven, Belgium
| | - Elisabeth Marcos
- Inserm U955 and Département de Physiologie, Hôpital Henri Mondor, Créteil, France, Université Paris-Est Créteil (UPEC), France
| | - Nathalie Mouraret
- Inserm U955 and Département de Physiologie, Hôpital Henri Mondor, Créteil, France, Université Paris-Est Créteil (UPEC), France
| | - Aurélien Parpaleix
- Inserm U955 and Département de Physiologie, Hôpital Henri Mondor, Créteil, France, Université Paris-Est Créteil (UPEC), France
| | - Régis Bobe
- Université Paris-Sud, Unité mixte de Recherche en Santé 770, Le Kremlin-Bicêtre, France
| | - Guillaume Gary-Bobo
- Inserm U955 and Département de Physiologie, Hôpital Henri Mondor, Créteil, France, Université Paris-Est Créteil (UPEC), France
| | - Mirna Saker
- Inserm U955 and Département de Physiologie, Hôpital Henri Mondor, Créteil, France, Université Paris-Est Créteil (UPEC), France
| | - Jean-Luc Dubois-Randé
- Service de Cardiologie, Hôpital Henri Mondor, AP-HP, 94010, Créteil, France; Université Paris-Est Créteil (UPEC)
| | - Mark T Gladwin
- Division of Pulmonary, Allergy and Critical Care Medicine, UPMC, Pittsburgh, PA
| | - Karen A Norris
- Heart, Lung, Blood and Vascular, University of Pittsburgh, Pittsburgh, PA
| | - Marion Delcroix
- Respiratory Division, University Hospitals of Leuven and Department of Clinical and Experimental Medicine, University of Leuven, Belgium
| | - Christophe Combadière
- Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France.,Inserm, U1135, CIMI-Paris, 91 Bd de l'hôpital, F-75013, Paris, France.,CNRS, ERL 8255, CIMI-Paris, 91 Bd de l'hôpital, F-75013, Paris, France
| | - Serge Adnot
- Inserm U955 and Département de Physiologie, Hôpital Henri Mondor, Créteil, France, Université Paris-Est Créteil (UPEC), France
| |
Collapse
|
44
|
Nogueira-Ferreira R, Ferreira R, Henriques-Coelho T. Cellular interplay in pulmonary arterial hypertension: Implications for new therapies. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:885-93. [DOI: 10.1016/j.bbamcr.2014.01.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 01/23/2014] [Accepted: 01/24/2014] [Indexed: 12/22/2022]
|
45
|
Zhang B, Niu W, Xu D, Li Y, Liu M, Wang Y, Luo Y, Zhao P, Liu Y, Dong M, Sun R, Dong H, Li Z. Oxymatrine prevents hypoxia- and monocrotaline-induced pulmonary hypertension in rats. Free Radic Biol Med 2014; 69:198-207. [PMID: 24440469 DOI: 10.1016/j.freeradbiomed.2014.01.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 01/07/2014] [Accepted: 01/08/2014] [Indexed: 02/02/2023]
Abstract
Pulmonary hypertension is a progressive disease characterized by marked pulmonary arterial remodeling and increased vascular resistance. Inflammation and oxidative stress promote the development of pulmonary hypertension. Oxymatrine, one of the main active components of the Chinese herb Sophora flavescens Ait. (Kushen), plays anti-inflammatory and antioxidant protective roles, which effects on pulmonary arteries remain unclear. This study aimed to investigate the effects of oxymatrine on pulmonary hypertension development. Sprague-Dawley rats were exposed to hypoxia for 28 days or injected with monocrotaline, to develop pulmonary hypertension, along with administration of oxymatrine (50mg/kg/day). Hemodynamics and pulmonary arterial remodeling data from the rats were then obtained. The antiproliferative effect of oxymatrine was verified by in vitro assays. The inflammatory cytokine mRNA levels and leukocyte and T cell accumulation in lung tissue were detected. The antioxidative effects of oxymatrine were explored in vitro. Our study shows that oxymatrine treatment attenuated right-ventricular systolic pressure and pulmonary arterial remodeling induced by hypoxia or monocrotaline and inhibited proliferation of pulmonary arterial smooth muscle cells (PASMCs). Increased expression of inflammatory cytokine mRNA and accumulation of leukocytes and T cells around the pulmonary arteries were suppressed with oxymatrine administration. Under hypoxic conditions, oxymatrine significantly upregulated Nrf2 and antioxidant protein SOD1 and HO-1 expression, but downregulated hydroperoxide levels in PASMCs. In summary, this study indicates that oxymatrine may prevent pulmonary hypertension through its antiproliferative, anti-inflammatory, and antioxidant effects, thus providing a promising pharmacological avenue for treating pulmonary hypertension.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Pathology, Xijing Hospital and Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, People's Republic of China.
| | - Wen Niu
- Department of Pathology, Xijing Hospital and Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Dunquan Xu
- Department of Pathology, Xijing Hospital and Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Yanyan Li
- Department of Pathology, Xijing Hospital and Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Manling Liu
- Department of Pathology, Xijing Hospital and Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Yanxia Wang
- Department of Pathology, Xijing Hospital and Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Ying Luo
- Department of Pathology, Xijing Hospital and Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Pengtao Zhao
- Department of Pathology, Xijing Hospital and Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Yi Liu
- Department of Pathology, Xijing Hospital and Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Mingqing Dong
- Department of Pathology, Xijing Hospital and Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Rihe Sun
- Xi'an YiLe Bio-Tech Laboratory, Xi'an 710075, People's Republic of China
| | - Haiying Dong
- Department of Pathology, Xijing Hospital and Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, People's Republic of China.
| | - Zhichao Li
- Department of Pathology, Xijing Hospital and Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, People's Republic of China.
| |
Collapse
|
46
|
El Chami H, Hassoun PM. Inflammatory mechanisms in the pathogenesis of pulmonary arterial hypertension. Compr Physiol 2013; 1:1929-41. [PMID: 23733693 DOI: 10.1002/cphy.c100028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inflammation is a prominent feature of human and experimental pulmonary hypertension (PH) as suggested by infiltration of various inflammatory cells and increased expression of certain cytokines in remodeled pulmonary vessels. Macrophages, T and B lymphocytes, and dendritic cells are found in the vascular lesions of idiopathic pulmonary arterial hypertension (PAH) as well as in PAH associated with connective tissue diseases or infectious etiologies such as HIV. In addition, PAH is often characterized by the presence of circulating chemokines and cytokines, increased expression of growth (such as VEGF and PDGF) and transcriptional (e.g., nuclear factor of activated T cells or NFAT) factors, and viral protein components (e.g., HIV-1 Nef), which directly contribute to further recruitment of inflammatory cells and the pulmonary vascular remodeling process. These inflammatory pathways may thus serve as potential specific therapeutic targets. This article provides an overview of inflammatory pathways involving chemokines and cytokines as well as growth factors, highlighting their potential role in pulmonary vascular remodeling and the possibility of future targeted therapy.
Collapse
Affiliation(s)
- Hala El Chami
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | | |
Collapse
|
47
|
Gomberg-Maitland M, Bull TM, Saggar R, Barst RJ, Elgazayerly A, Fleming TR, Grimminger F, Rainisio M, Stewart DJ, Stockbridge N, Ventura C, Ghofrani AH, Rubin LJ. New trial designs and potential therapies for pulmonary artery hypertension. J Am Coll Cardiol 2013; 62:D82-D91. [PMID: 24355645 PMCID: PMC4117578 DOI: 10.1016/j.jacc.2013.10.026] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 10/22/2013] [Indexed: 11/25/2022]
Abstract
A greater understanding of the epidemiology, pathogenesis, and pathophysiology of pulmonary artery hypertension (PAH) has led to significant advances, but the disease remains fatal. Treatment options are neither universally available nor always effective, underscoring the need for development of novel therapies and therapeutic strategies. Clinical trials to date have provided evidence of efficacy, but were limited in evaluating the scope and duration of treatment effects. Numerous potential targets in varied stages of drug development exist, in addition to novel uses of familiar therapies. The pursuit of gene and cell-based therapy continues, and device use to help acute deterioration and chronic management is emerging. This rapid surge of drug development has led to multicenter pivotal clinical trials and has resulted in novel ethical and global clinical trial concerns. This paper will provide an overview of the opportunities and challenges that await the development of novel treatments for PAH.
Collapse
Affiliation(s)
- Mardi Gomberg-Maitland
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, Illinois.
| | - Todd M Bull
- Section of Pulmonary and Critical Care, Department of Medicine, University of Colorado, Aurora, Colorado
| | | | | | | | - Thomas R Fleming
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Friedrich Grimminger
- Section of Pulmonary, Department of Medicine, Department of Medical Oncology, University Hospital Giessen, Giessen, Germany
| | | | - Duncan J Stewart
- Section of Cardiology, Department of Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | | | - Carlo Ventura
- Section of Cardiology, Department of Medicine, University of Bologna, Bologna, Italy
| | - Ardeschir H Ghofrani
- Section of Pulmonary, Department of Medicine, Department of Medical Oncology, University Hospital Giessen, Giessen, Germany
| | - Lewis J Rubin
- Section of Pulmonary and Critical Care, Department of Medicine, University of California San Diego, San Diego, California
| |
Collapse
|
48
|
Tajsic T, Morrell NW. Smooth muscle cell hypertrophy, proliferation, migration and apoptosis in pulmonary hypertension. Compr Physiol 2013; 1:295-317. [PMID: 23737174 DOI: 10.1002/cphy.c100026] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Pulmonary hypertension is a multifactorial disease characterized by sustained elevation of pulmonary vascular resistance (PVR) and pulmonary arterial pressure (PAP). Central to the pathobiology of this disease is the process of vascular remodelling. This process involves structural and functional changes to the normal architecture of the walls of pulmonary arteries (PAs) that lead to increased muscularization of the muscular PAs, muscularization of the peripheral, previously nonmuscular, arteries of the respiratory acinus, formation of neointima, and formation of plexiform lesions. Underlying or contributing to the development of these lesions is hypertrophy, proliferation, migration, and resistance to apoptosis of medial cells and this article is concerned with the cellular and molecular mechanisms of these processes. In the first part of the article we focus on the concept of smooth muscle cell phenotype and the difficulties surrounding the identification and characterization of the cell/cells involved in the remodelling of the vessel media and we review the general mechanisms of cell hypertrophy, proliferation, migration and apoptosis. Then, in the larger part of the article, we review the factors identified thus far to be involved in PH intiation and/or progression and review and discuss their effects on pulmonary artery smooth muscle cells (PASMCs) the predominant cells in the tunica media of PAs.
Collapse
Affiliation(s)
- Tamara Tajsic
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | | |
Collapse
|
49
|
Kherbeck N, Tamby MC, Bussone G, Dib H, Perros F, Humbert M, Mouthon L. The role of inflammation and autoimmunity in the pathophysiology of pulmonary arterial hypertension. Clin Rev Allergy Immunol 2013; 44:31-8. [PMID: 21394427 DOI: 10.1007/s12016-011-8265-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Pulmonary arterial hypertension is characterized by a remodeling of pulmonary arteries with endothelial cell, fibroblast, and vascular smooth muscle cell activation and proliferation. Since pulmonary arterial hypertension occurs frequently in autoimmune conditions such as systemic sclerosis, inflammation and autoimmunity have been suspected to play a critical role in both idiopathic pulmonary arterial hypertension and systemic sclerosis-associated pulmonary arterial hypertension. High levels of pro-inflammatory cytokines such as interleukin-1 and interleukin-6, platelet-derived growth factor, or macrophage inflammatory protein 1 have been found in lung samples of patients with pulmonary arterial hypertension, along with inflammatory cell infiltrates mainly composed of macrophages and dendritic cells, T and B lymphocytes. In addition, circulating autoantibodies are found in the peripheral blood of patients. Thus, autoimmunity and inflammation probably play a role in the development of pulmonary arterial hypertension. In this setting, it would be important to set-up new experimental models of pulmonary arterial hypertension, in order to define novel therapeutics that specifically target immune disturbances in this devastating condition.
Collapse
|
50
|
Role of Rho-kinase and its inhibitors in pulmonary hypertension. Pharmacol Ther 2013; 137:352-64. [DOI: 10.1016/j.pharmthera.2012.12.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 10/27/2012] [Indexed: 11/20/2022]
|