1
|
Habibi A, Ruf W, Schurgers L. Protease-activated receptors in vascular smooth muscle cells: a bridge between thrombo-inflammation and vascular remodelling. Cell Commun Signal 2025; 23:57. [PMID: 39891111 PMCID: PMC11786455 DOI: 10.1186/s12964-025-02066-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/25/2025] [Indexed: 02/03/2025] Open
Abstract
Coagulation factors are responsible for blood clot formation yet have also non-canonical functions as signalling molecules. In this context, they can activate protease-activated receptors (PARs) ubiquitously expressed in the vasculature. During vascular repair, vascular smooth muscle cells (VSMCs) will switch from a contractile to a synthetic reparative phenotype. During prolonged vascular stress, VSMCs acquire a pathological phenotype leading to cardiovascular disease. Activated coagulation factors impact on vessel wall permeability and integrity after vascular injury with a key role for PAR activation on endothelial cells. The activation of PARs on VSMCs supports vessel wall repair following injury. Prolonged PAR activation, however, results in pathological vascular remodelling. Therefore, understanding the mechanisms of PAR activation on VSMCs is key to propel our understanding of the molecular and cellular mechanisms to develop novel therapeutic strategies to resolve vascular remodelling.In this review, we discuss recent advances on the role of PAR signalling on VSMCs and specifically their role in vascular remodelling contributing to cardiovascular disease. Additionally, we discuss current therapeutic strategies targeting PAR signalling - indirectly or directly - in relation to cardiovascular disease.
Collapse
Affiliation(s)
- Anxhela Habibi
- Department of Biochemistry, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands.
- Center for Thrombosis and Hemostasis, Johannes-Gutenberg-University Medical Center Mainz, Mainz, Germany.
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes-Gutenberg-University Medical Center Mainz, Mainz, Germany
| | - Leon Schurgers
- Department of Biochemistry, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
2
|
Harr TJ, Gupta N, Rahar B, Stott K, Medina-Guevara Y, Gari MK, Oler AT, McDermott IS, Lee HJ, Rasoulianboroujeni M, Weichmann AM, Forati A, Holbert K, Langel TS, Coulter KW, Burkel BM, Tomasini-Johansson BR, Ponik SM, Engle JW, Hernandez R, Kwon GS, Sandbo N, Bernau K. The fibronectin-targeting PEG-FUD imaging probe shows enhanced uptake during fibrogenesis in experimental lung fibrosis. Respir Res 2025; 26:34. [PMID: 39844185 PMCID: PMC11756063 DOI: 10.1186/s12931-025-03107-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
Progressive forms of interstitial lung diseases, including idiopathic pulmonary fibrosis (IPF), are deadly disorders lacking non-invasive biomarkers for assessment of early disease activity, which presents a major obstacle in disease management. Excessive extracellular matrix (ECM) deposition is a hallmark of these disorders, with fibronectin being an abundant ECM glycoprotein that is highly upregulated in early fibrosis and serves as a scaffold for the deposition of other matrix proteins. Due to its role in active fibrosis, we are targeting fibronectin as a biomarker of early lung fibrosis disease activity via the PEGylated fibronectin-binding polypeptide (PEG-FUD). In this work, we demonstrate the binding of PEG-FUD to the fibrotic lung throughout the course of bleomycin-induced murine model of pulmonary fibrosis. We first analyzed the binding of radiolabeled PEG-FUD following direct incubation to precision cut lung slices from mice at different stages of experimental lung fibrosis. Then, we administered fluorescently labeled PEG-FUD subcutaneously to mice over the course of bleomycin-induced pulmonary fibrosis and assessed peptide uptake 24 h later through ex vivo tissue imaging. Using both methods, we found that peptide targeting to the fibrotic lung is increased during the fibrogenic phase of the single dose bleomycin lung fibrosis model (days 7 and 14 post-bleomycin). At these timepoints we found a correlative relationship between peptide uptake and fibrotic burden. These data suggest that PEG-FUD targets fibronectin associated with active fibrogenesis in this model, making it a promising candidate for a clinically translatable molecular imaging probe to non-invasively determine pulmonary fibrosis disease activity, enabling accelerated therapeutic decision-making.
Collapse
Affiliation(s)
- Thomas J Harr
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Nikesh Gupta
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, 53705, USA
| | - Babita Rahar
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Kristen Stott
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Yadira Medina-Guevara
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Metti K Gari
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Angie T Oler
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Ivy Sohee McDermott
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Hye Jin Lee
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, 53705, USA
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Morteza Rasoulianboroujeni
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, 53705, USA
| | - Ashley M Weichmann
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, USA
| | - Amir Forati
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin- Madison, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Kelsey Holbert
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Trevor S Langel
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Kade W Coulter
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Brian M Burkel
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Bianca R Tomasini-Johansson
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, USA
| | - Jonathan W Engle
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - Reinier Hernandez
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, USA
| | - Glen S Kwon
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, USA
| | - Nathan Sandbo
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Ksenija Bernau
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI, 53792, USA.
| |
Collapse
|
3
|
Cenerini G, Chimera D, Pagnini M, Bazzan E, Conti M, Turato G, Celi A, Neri T. The Intricate Relationship Between Pulmonary Fibrosis and Thrombotic Pathology: A Narrative Review. Cells 2024; 13:2099. [PMID: 39768190 PMCID: PMC11674501 DOI: 10.3390/cells13242099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is associated with a significantly increased risk of thrombotic events and mortality. This review explores the complex bidirectional relationship between pulmonary fibrosis and thrombosis, discussing epidemiological evidence, pathogenetic mechanisms, and therapeutic implications, with a particular focus on the emerging role of extracellular vesicles (EVs) as crucial mediators linking fibrosis and coagulation. Coagulation factors directly promote fibrosis, while fibrosis itself activates thrombotic pathways. Retrospective studies suggest the benefits of anticoagulants in IPF, but prospective trials have faced challenges. Novel anticoagulants, profibrinolytic therapies, and agents targeting protease-activated receptors (PARs) show promise in preclinical studies and early clinical trials. EVs have emerged as key players in the pathogenesis of interstitial lung diseases (ILDs), serving as vehicles for intercellular communication and contributing to both fibrosis and coagulation. EV-based approaches, such as EV modulation, engineered EVs as drug delivery vehicles, and mesenchymal stem cell-derived EVs, represent promising therapeutic strategies. Ongoing research should focus on optimizing risk-benefit profiles, identifying predictive biomarkers, evaluating combination strategies targeting thrombotic, fibrotic, and inflammatory pathways, and advancing the understanding of EVs in ILDs to develop targeted interventions.
Collapse
Affiliation(s)
- Giovanni Cenerini
- UO Pneumologia, Azienda Ospedaliero-Universitaria Pisana, 56124 Pisa, Italy; (G.C.); (D.C.)
| | - Davide Chimera
- UO Pneumologia, Azienda Ospedaliero-Universitaria Pisana, 56124 Pisa, Italy; (G.C.); (D.C.)
| | - Marta Pagnini
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università degli Studi di Pisa, 56124 Pisa, Italy; (M.P.); (T.N.)
| | - Erica Bazzan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy; (E.B.); (M.C.); (G.T.)
| | - Maria Conti
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy; (E.B.); (M.C.); (G.T.)
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Graziella Turato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, 35128 Padova, Italy; (E.B.); (M.C.); (G.T.)
| | - Alessandro Celi
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università degli Studi di Pisa, 56124 Pisa, Italy; (M.P.); (T.N.)
| | - Tommaso Neri
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università degli Studi di Pisa, 56124 Pisa, Italy; (M.P.); (T.N.)
| |
Collapse
|
4
|
Li X, Liu Y, Tang Y, Xia Z. Transformation of macrophages into myofibroblasts in fibrosis-related diseases: emerging biological concepts and potential mechanism. Front Immunol 2024; 15:1474688. [PMID: 39386212 PMCID: PMC11461261 DOI: 10.3389/fimmu.2024.1474688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024] Open
Abstract
Macrophage-myofibroblast transformation (MMT) transforms macrophages into myofibroblasts in a specific inflammation or injury microenvironment. MMT is an essential biological process in fibrosis-related diseases involving the lung, heart, kidney, liver, skeletal muscle, and other organs and tissues. This process consists of interacting with various cells and molecules and activating different signal transduction pathways. This review deeply discussed the molecular mechanism of MMT, clarified crucial signal pathways, multiple cytokines, and growth factors, and formed a complex regulatory network. Significantly, the critical role of transforming growth factor-β (TGF-β) and its downstream signaling pathways in this process were clarified. Furthermore, we discussed the significance of MMT in physiological and pathological conditions, such as pulmonary fibrosis and cardiac fibrosis. This review provides a new perspective for understanding the interaction between macrophages and myofibroblasts and new strategies and targets for the prevention and treatment of MMT in fibrotic diseases.
Collapse
Affiliation(s)
- Xiujun Li
- Health Science Center, Chifeng University, Chifeng, China
| | - Yuyan Liu
- Rehabilitation Medicine College, Shandong Second Medical University, Jinan, China
| | - Yongjun Tang
- Department of Emergency, Affiliated Hospital of Chifeng University, Chifeng, China
| | - Zhaoyi Xia
- Department of Library, Children’s Hospital Affiliated to Shandong University, Jinan, China
- Department of Library, Jinan Children’s Hospital, Jinan, China
| |
Collapse
|
5
|
Chia ZJ, Cao YN, Little PJ, Kamato D. Transforming growth factor-β receptors: versatile mechanisms of ligand activation. Acta Pharmacol Sin 2024; 45:1337-1348. [PMID: 38351317 PMCID: PMC11192764 DOI: 10.1038/s41401-024-01235-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/28/2024] [Indexed: 02/19/2024]
Abstract
Transforming growth factor-β (TGF-β) signaling is initiated by activation of transmembrane TGF-β receptors (TGFBR), which deploys Smad2/3 transcription factors to control cellular responses. Failure or dysregulation in the TGF-β signaling pathways leads to pathological conditions. TGF-β signaling is regulated at different levels along the pathways and begins with the liberation of TGF-β ligand from its latent form. The mechanisms of TGFBR activation display selectivity to cell types, agonists, and TGF-β isoforms, enabling precise control of TGF-β signals. In addition, the cell surface compartments used to release active TGF-β are surprisingly vibrant, using thrombospondins, integrins, matrix metalloproteinases and reactive oxygen species. The scope of TGFBR activation is further unfolded with the discovery of TGFBR activation initiated by other signaling pathways. The unique combination of mechanisms works in series to trigger TGFBR activation, which can be explored as therapeutic targets. This comprehensive review provides valuable insights into the diverse mechanisms underpinning TGFBR activation, shedding light on potential avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Zheng-Jie Chia
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia
| | - Ying-Nan Cao
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia.
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia.
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia.
| |
Collapse
|
6
|
Dai C, Lin X, Qi Y, Wang Y, Lv Z, Zhao F, Deng Z, Feng X, Zhang T, Pu X. Vitamin D3 improved hypoxia-induced lung injury by inhibiting the complement and coagulation cascade and autophagy pathway. BMC Pulm Med 2024; 24:9. [PMID: 38166725 PMCID: PMC10759436 DOI: 10.1186/s12890-023-02784-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Pulmonary metabolic dysfunction can cause lung tissue injury. There is still no ideal drug to protect against hypoxia-induced lung injury, therefore, the development of new drugs to prevent and treat hypoxia-induced lung injury is urgently needed. We aimed to explore the ameliorative effects and molecular mechanisms of vitamin D3 (VD3) on hypoxia-induced lung tissue injury. METHODS Sprague-Dawley (SD) rats were randomly divided into three groups: normoxia, hypoxia, and hypoxia + VD3. The rat model of hypoxia was established by placing the rats in a hypobaric chamber. The degree of lung injury was determined using hematoxylin and eosin (H&E) staining, lung water content, and lung permeability index. Transcriptome data were subjected to differential gene expression and pathway analyses. In vitro, type II alveolar epithelial cells were co-cultured with hepatocytes and then exposed to hypoxic conditions for 24 h. For VD3 treatment, the cells were treated with low and high concentrations of VD3. RESULTS Transcriptome and KEGG analyses revealed that VD3 affects the complement and coagulation cascade pathways in hypoxia-induced rats, and the genes enriched in this pathway were Fgb/Fga/LOC100910418. Hypoxia can cause increases in lung edema, inflammation, and lung permeability disruption, which are attenuated by VD3 treatment. VD3 weakened the complement and coagulation cascade in the lung and liver of hypoxia-induced rats, characterized by lower expression of fibrinogen alpha chain (Fga), fibrinogen beta chain (Fgb), protease-activated receptor 1 (PAR1), protease-activated receptor 3 (PAR3), protease-activated receptor 4 (PAR4), complement (C) 3, C3a, and C5. In addition, VD3 improved hypoxic-induced type II alveolar epithelial cell damage and inflammation by inhibiting the complement and coagulation cascades. Furthermore, VD3 inhibited hypoxia-induced autophagy in vivo and in vitro, which was abolished by the mitophagy inducer, carbonyl cyanide-m-chlorophenylhydrazone (CCCP). CONCLUSION VD3 alleviated hypoxia-induced pulmonary edema by inhibiting the complement and coagulation cascades and autophagy pathways.
Collapse
Affiliation(s)
- Chongyang Dai
- Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Xue Lin
- West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610000, People's Republic of China
| | - Yinglian Qi
- Qinghai Normal University, Xining, Qinghai Province, 810008, People's Republic of China
| | - Yaxuan Wang
- Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Zhongkui Lv
- Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Fubang Zhao
- Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Zhangchang Deng
- Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China
| | - Xiaokai Feng
- Department of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China.
- Department of Respiratory and Critical Care Medicine, Qinghai Provincial People's Hospital, Qinghai University, Xining, Qinghai Province, 810007, People's Republic of China.
| | - Tongzuo Zhang
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai Province, 810001, People's Republic of China.
| | - Xiaoyan Pu
- Qinghai University, Xining, Qinghai Province, 810016, People's Republic of China.
| |
Collapse
|
7
|
Heidari Z, Naeimzadeh Y, Fallahi J, Savardashtaki A, Razban V, Khajeh S. The Role of Tissue Factor In Signaling Pathways of Pathological Conditions and Angiogenesis. Curr Mol Med 2024; 24:1135-1151. [PMID: 37817529 DOI: 10.2174/0115665240258746230919165935] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/10/2023] [Accepted: 07/27/2023] [Indexed: 10/12/2023]
Abstract
Tissue factor (TF) is an integral transmembrane protein associated with the extrinsic coagulation pathway. TF gene expression is regulated in response to inflammatory cytokines, bacterial lipopolysaccharides, and mechanical injuries. TF activity may be affected by phosphorylation of its cytoplasmic domain and alternative splicing. TF acts as the primary initiator of physiological hemostasis, which prevents local bleeding at the injury site. However, aberrant expression of TF, accompanied by the severity of diseases and infections under various pathological conditions, triggers multiple signaling pathways that support thrombosis, angiogenesis, inflammation, and metastasis. Protease-activated receptors (PARs) are central in the downstream signaling pathways of TF. In this study, we have reviewed the TF signaling pathways in different pathological conditions, such as wound injury, asthma, cardiovascular diseases (CVDs), viral infections, cancer and pathological angiogenesis. Angiogenic activities of TF are critical in the repair of wound injuries and aggressive behavior of tumors, which are mainly performed by the actions of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1 (HIF1-α). Pro-inflammatory effects of TF have been reported in asthma, CVDs and viral infections, including COVID-19, which result in tissue hypertrophy, inflammation, and thrombosis. TF-FVII induces angiogenesis via clotting-dependent and -independent mechanisms. Clottingdependent angiogenesis is induced via the generation of thrombin and cross-linked fibrin network, which facilitate vessel infiltration and also act as a reservoir for endothelial cells (ECs) growth factors. Expression of TF in tumor cells and ECs triggers clotting-independent angiogenesis through induction of VEGF, urokinase-type plasminogen activator (uPAR), early growth response 1 (EGR1), IL8, and cysteine-rich angiogenic inducer 61 (Cyr61).
Collapse
Affiliation(s)
- Zahra Heidari
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sahar Khajeh
- Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
8
|
Ghosh K, Shome DK, Kulkarni B, Ghosh MK, Ghosh K. Fibrosis and bone marrow: understanding causation and pathobiology. J Transl Med 2023; 21:703. [PMID: 37814319 PMCID: PMC10561412 DOI: 10.1186/s12967-023-04393-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/28/2023] [Indexed: 10/11/2023] Open
Abstract
Bone marrow fibrosis represents an important structural change in the marrow that interferes with some of its normal functions. The aetiopathogenesis of fibrosis is not well established except in its primary form. The present review consolidates current understanding of marrow fibrosis. We searched PubMed without time restriction using key words: bone marrow and fibrosis as the main stem against the terms: growth factors, cytokines and chemokines, morphology, megakaryocytes and platelets, myeloproliferative disorders, myelodysplastic syndrome, collagen biosynthesis, mesenchymal stem cells, vitamins and minerals and hormones, and mechanism of tissue fibrosis. Tissue marrow fibrosis-related papers were short listed and analysed for the review. It emerged that bone marrow fibrosis is the outcome of complex interactions between growth factors, cytokines, chemokines and hormones together with their facilitators and inhibitors. Fibrogenesis is initiated by mobilisation of special immunophenotypic subsets of mesenchymal stem cells in the marrow that transform into fibroblasts. Fibrogenic stimuli may arise from neoplastic haemopoietic or non-hematopoietic cells, as well as immune cells involved in infections and inflammatory conditions. Autoimmunity is involved in a small subset of patients with marrow fibrosis. Megakaryocytes and platelets are either directly involved or are important intermediaries in stimulating mesenchymal stem cells. MMPs, TIMPs, TGF-β, PDGRF, and basic FGF and CRCXL4 chemokines are involved in these processes. Genetic and epigenetic changes underlie many of these conditions.
Collapse
Affiliation(s)
- Kanjaksha Ghosh
- National Institute of Immunohaematology, 13 Th Fl KEM Hospital, Parel, Mumbai, 400012, India.
| | - Durjoy K Shome
- Department of Pathophysiology, American University of Antigua College of Medicine, Coolidge, Antigua and Barbuda
| | - Bipin Kulkarni
- Department of Molecular Biology and Haemostasis, National Institute of Immunohaematology, 13Th Fl KEM Hospital, Parel, Mumbai, 400012, India
| | - Malay K Ghosh
- Department of Haematology, Nilratan Sarkar Medical College, Kolkata, 700014, West Bengal, India
| | - Kinjalka Ghosh
- Department of Clinical Biochemistry, Tata Medical Centre and Homi Bhaba National Institute, Parel, Mumbai, 400012, India
| |
Collapse
|
9
|
Liu Q, Liu X, Wang G, Wu F, Hou Y, Liu H. Genome-wide DNA methylation analysis of Astragalus and Danshen on the intervention of myofibroblast activation in idiopathic pulmonary fibrosis. BMC Pulm Med 2023; 23:325. [PMID: 37667288 PMCID: PMC10478235 DOI: 10.1186/s12890-023-02601-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/09/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF), a chronic progressive interstitial lung disease of unknown etiology, is characterized by continuous damage to alveolar epithelial cells, abnormal repair of alveolar tissue, and alveolar wall scar formation. Currently, the recommended treatment for IPF in Western medicine is relatively limited. In contrast, traditional Chinese medicine and compound prescriptions show advantages in the diagnosis and treatment of IPF, which can be attributed to their multi-channel and multi-target characteristics and minimal side-effects. The purpose of this study was to further corroborate the effectiveness and significance of the traditional Chinese medications Astragalus and Danshen in IPF treatment. METHODS We performed whole-genome methylation analysis on nine rat lung tissue samples to determine the epigenetic variation between IPF and non-fibrotic lungs using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses and quantitative reverse transcription polymerase chain reactions. RESULTS We identified differentially methylated regions and 105 associated key functional genes in samples related to IPF and Chinese medicine treatment. Based on the methylation levels and gene expression profiles between the Chinese medicine intervention and pulmonary fibrosis model groups, we speculated that Astragalus and Salvia miltiorrhiza (traditionally known as Danshen) act on the Isl1, forkhead box O3, and Sonic hedgehog genes via regulation at transcriptional and epigenetic levels during IPF. CONCLUSIONS These findings provide novel insights into the epigenetic regulation of IPF, indicate the effectiveness of Astragalus and Danshen in treating IPF, and suggest several promising therapeutic targets for preventing and treating IPF.
Collapse
Affiliation(s)
- Qingyin Liu
- Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, University Science Park, Changqing District, Jinan City, 250355, China
| | - Xue Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369, Jing Shi Road, Jinan City, 250013, China
| | - Guoyu Wang
- Capital Medical University, No. 10, Xizhang Road, Youanmenwai, Fengtai District, Beijing, 100069, China
| | - Fan Wu
- Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, University Science Park, Changqing District, Jinan City, 250355, China
| | - Yuan Hou
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369, Jing Shi Road, Jinan City, 250013, China
| | - Huaman Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369, Jing Shi Road, Jinan City, 250013, China.
| |
Collapse
|
10
|
Poe A, Martinez Yus M, Wang H, Santhanam L. Lysyl oxidase like-2 in fibrosis and cardiovascular disease. Am J Physiol Cell Physiol 2023; 325:C694-C707. [PMID: 37458436 PMCID: PMC10635644 DOI: 10.1152/ajpcell.00176.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 09/01/2023]
Abstract
Fibrosis is an important and essential reparative response to injury that, if left uncontrolled, results in the excessive synthesis, deposition, remodeling, and stiffening of the extracellular matrix, which is deleterious to organ function. Thus, the sustained activation of enzymes that catalyze matrix remodeling and cross linking is a fundamental step in the pathology of fibrotic diseases. Recent studies have implicated the amine oxidase lysyl oxidase like-2 (LOXL2) in this process and established significantly elevated expression of LOXL2 as a key component of profibrotic conditions in several organ systems. Understanding the relationship between LOXL2 and fibrosis as well as the mechanisms behind these relationships can offer significant insights for developing novel therapies. Here, we summarize the key findings that demonstrate the link between LOXL2 and fibrosis and inflammation, examine current therapeutics targeting LOXL2 for the treatment of fibrosis, and discuss future directions for experiments and biomedical engineering.
Collapse
Affiliation(s)
- Alan Poe
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
| | - Marta Martinez Yus
- Department of Anesthesiology and CCM, Johns Hopkins University, Baltimore, Maryland, United States
| | - Huilei Wang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
| | - Lakshmi Santhanam
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Anesthesiology and CCM, Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
11
|
Wei J, Zhan J, Ji H, Xu Y, Xu Q, Zhu X, Liu Y. Fibroblast Upregulation of Vitamin D Receptor Represents a Self-Protective Response to Limit Fibroblast Proliferation and Activation during Pulmonary Fibrosis. Antioxidants (Basel) 2023; 12:1634. [PMID: 37627629 PMCID: PMC10451996 DOI: 10.3390/antiox12081634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Dysregulation of vitamin D receptor (VDR) is implicated in chronic obstructive pulmonary disease. However, whether VDR dysregulation contributes to the development of pulmonary fibrosis remains largely unknown. Analysis of bulk and single-cell RNA profiling datasets revealed VDR upregulation in lung fibroblasts from patients with pulmonary fibrosis or fibrotic mice, which was validated in lung fibroblasts from bleomycin-exposed mice and bleomycin-treated fibroblasts. Stable VDR knockdown promoted, whereas the VDR agonist paricalcitol suppressed lung fibroblast proliferation and activation. Gene set enrichment analysis (GSEA) showed that the JAK/STAT pathway and unfolded protein response (UPR), a process related to endoplasmic reticulum (ER) stress, were enriched in lung fibroblasts of fibrotic lungs. Stable VDR knockdown stimulated, but paricalcitol suppressed ER stress and JAK1/STAT3 activation in lung fibroblasts. The STAT3 inhibitor blocked bleomycin- or stable VDR knockdown-induced ER stress. Paricalcitol inhibited the bleomycin-induced enrichment of STAT3 to the ATF6 promoter, thereby suppressing ATF6 expression in fibroblasts. Paricalcitol or intrapulmonary VDR overexpression inactivated JAK1/STAT3 and suppressed ER stress in bleomycin-treated mice, thus resulting in the inhibition of fibroblast proliferation and activation. Collectively, this study suggests that fibroblast VDR upregulation may be a self-protective response to limit fibroblast proliferation and activation during pulmonary fibrosis by suppressing the JAK1/STAT3/ER stress pathway.
Collapse
Affiliation(s)
- Juan Wei
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
- School of Sports and Health, Nanjing Sport Institute, Nanjing 210014, China
| | - Junhui Zhan
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| | - Hui Ji
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| | - Yitong Xu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| | - Qingfeng Xu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| | - Xiaoyan Zhu
- Department of Physiology, Navy Medical University, Shanghai 200433, China
| | - Yujian Liu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (J.Z.); (H.J.); (Y.X.); (Q.X.)
| |
Collapse
|
12
|
Wu C, Zhong Q, Shrestha R, Wang J, Hu X, Li H, Rouchka EC, Yan J, Ding C. Reactive myelopoiesis and FX-expressing macrophages triggered by chemotherapy promote cancer lung metastasis. JCI Insight 2023; 8:e167499. [PMID: 36976637 PMCID: PMC10243818 DOI: 10.1172/jci.insight.167499] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Several preclinical studies have demonstrated that certain cytotoxic drugs enhance metastasis, but the importance of host responses triggered by chemotherapy in regulating cancer metastasis has not been fully explored. Here, we showed that multidose gemcitabine (GEM) treatment promoted breast cancer lung metastasis in a transgenic spontaneous breast cancer model. GEM treatment significantly increased accumulation of CCR2+ macrophages and monocytes in the lungs of tumor-bearing as well as tumor-free mice. These changes were largely caused by chemotherapy-induced reactive myelopoiesis biased toward monocyte development. Mechanistically, enhanced production of mitochondrial ROS was observed in GEM-treated BM Lin-Sca1+c-Kit+ cells and monocytes. Treatment with the mitochondria targeted antioxidant abrogated GEM-induced hyperdifferentiation of BM progenitors. In addition, GEM treatment induced upregulation of host cell-derived CCL2, and knockout of CCR2 signaling abrogated the pro-metastatic host response induced by chemotherapy. Furthermore, chemotherapy treatment resulted in the upregulation of coagulation factor X (FX) in lung interstitial macrophages. Targeting activated FX (FXa) using FXa inhibitor or F10 gene knockdown reduced the pro-metastatic effect of chemotherapy. Together, these studies suggest a potentially novel mechanism for chemotherapy-induced metastasis via the host response-induced accumulation of monocytes/macrophages and interplay between coagulation and inflammation in the lungs.
Collapse
Affiliation(s)
- Caijun Wu
- UofL Health - Brown Cancer Center and
| | | | - Rejeena Shrestha
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | | | | | - Hong Li
- UofL Health - Brown Cancer Center and
| | - Eric C. Rouchka
- Department of Computer Science and Engineering, University of Louisville J.B. Speed School of Engineering, Louisville, Kentucky, USA
| | - Jun Yan
- UofL Health - Brown Cancer Center and
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
- Department of Surgery, Division of Immunotherapy, UofL Health - Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Chuanlin Ding
- UofL Health - Brown Cancer Center and
- Department of Surgery, Division of Immunotherapy, UofL Health - Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
13
|
Wang H, Poe A, Martinez Yus M, Pak L, Nandakumar K, Santhanam L. Lysyl oxidase-like 2 processing by factor Xa modulates its activity and substrate preference. Commun Biol 2023; 6:375. [PMID: 37029269 PMCID: PMC10082071 DOI: 10.1038/s42003-023-04748-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 03/23/2023] [Indexed: 04/09/2023] Open
Abstract
Lysyl oxidase-like 2 (LOXL2) has been identified as an essential mediator of extracellular matrix (ECM) remodeling in several disease processes including cardiovascular disease. Thus, there is growing interest in understanding the mechanisms by which LOXL2 is regulated in cells and tissue. While LOXL2 occurs both in full length and processed forms in cells and tissue, the precise identity of the proteases that process LOXL2 and the consequences of processing on LOXL2's function remain incompletely understood. Here we show that Factor Xa (FXa) is a protease that processes LOXL2 at Arg-338. Processing by FXa does not affect the enzymatic activity of soluble LOXL2. However, in situ in vascular smooth muscle cells, LOXL2 processing by FXa results in decreased cross-linking activity in the ECM and shifts substrate preference of LOXL2 from type IV collagen to type I collagen. Additionally, processing by FXa increases the interactions between LOXL2 and prototypical LOX, suggesting a potential compensatory mechanism to preserve total LOXs activity in the vascular ECM. FXa expression is prevalent in various organ systems and shares similar roles in fibrotic disease progression as LOXL2. Thus, LOXL2 processing by FXa could have significant implications in pathologies where LOXL2 is involved.
Collapse
Affiliation(s)
- Huilei Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD, 21205, USA
| | - Alan Poe
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD, 21205, USA
| | - Marta Martinez Yus
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, 3400 N Charles St, Baltimore, MD, 21218, USA
| | - Lydia Pak
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD, 21205, USA
| | - Kavitha Nandakumar
- Department of Anesthesiology and CCM, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore MD, 21205, Baltimore, MD, USA
| | - Lakshmi Santhanam
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD, 21205, USA.
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, 3400 N Charles St, Baltimore, MD, 21218, USA.
- Department of Anesthesiology and CCM, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore MD, 21205, Baltimore, MD, USA.
| |
Collapse
|
14
|
Sun H, Liu M, Yang X, Xi L, Xu W, Deng M, Ren Y, Xie W, Dai H, Wang C. Incidence and risk factors of venous thrombotic events in patients with interstitial lung disease during hospitalization. Thromb J 2023; 21:17. [PMID: 36765371 PMCID: PMC9912624 DOI: 10.1186/s12959-023-00458-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/27/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Studies on the incidence of venous thromboembolism (VTE) events in patients with interstitial lung disease (ILD) are limited and the results are inconsistent. The aim of this research was to investigate the incidence and risk factors of VTE in ILD during hospitalization. MATERIALS AND METHODS In this retrospective, cross-sectional, observational study, a total of 5009 patients diagnosed with ILD from January 2016 to March 2022 in our hospital were retrospectively included. In ILD patients, VTE including pulmonary thromboembolism (PTE) and deep vein thrombosis (DVT) were screened from the electronic medical record system. Diagnosis of PTE and DVT were performed by CT pulmonary angiography (CTPA), CTV or ultrasound. And then the incidence and risk factors of VTE in different types of ILD were assessed. RESULTS Among 5009 patients with ILD, VTE was detected in 129 (2.6%) patients, including 15(0.3%) patients with both PTE and DVT, 34 (0.7%) patients with PTE and 80 (1.6%) patients with DVT. 85.1% of patients with APE were in the intermediate-low risk group. The incidence of VTE in Anti-Neutrophil Cytoplasmic Antibodies -associated vasculitis related ILD (ANCA-AV-ILD), hypersensitivity pneumonitis and idiopathic pulmonary fibrosis (IPF) respectively was 7.9% and 3.6% and 3.5%. In patients with connective tissue disease-associated ILD (CTD-ILD), the incidence of VTE, DVT, PTE, combined PTE and DVT respectively was 3.0%, 2.3%, 0.4% and 0.3%. Among the various risk factors, different ILD categories, age ≥ 80 years (OR 4.178, 95% CI 2.097-8.321, P < 0.001), respiratory failure (OR 2.382, 95% CI 1.533-3.702, P < 0.001) and varicose veins (OR 3.718, 95% CI 1.066-12.964, P = 0.039) were independent risk factors of VTE. The incidence of VTE in patients with ILD increased with the length of time in hospital from 2.2% (< 7 days) to 6.4% (> 21 days). CONCLUSION The incidence of VTE during hospitalization in ILD patients was 2.6%, with a 1.6% incidence of DVT, higher than the 0.7% incidence of PTE. Advanced age, ILD categories, respiratory failure and varicose veins as independent risk factors for the development of VTE should be closely monitored.
Collapse
Affiliation(s)
- Haishuang Sun
- grid.430605.40000 0004 1758 4110Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, 130021 China ,National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, 100029 Beijing, China ,grid.506261.60000 0001 0706 7839Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Min Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Xiaoyan Yang
- National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, 100029 Beijing, China
| | - Linfeng Xi
- National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, 100029 Beijing, China
| | - Wenqing Xu
- grid.415954.80000 0004 1771 3349Department of Radiology, China-Japan Friendship Hospital, Beijing, 100029 China
| | - Mei Deng
- grid.506261.60000 0001 0706 7839Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China ,grid.415954.80000 0004 1771 3349Department of Radiology, China-Japan Friendship Hospital, Beijing, 100029 China
| | - Yanhong Ren
- National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, 100029 Beijing, China
| | - Wanmu Xie
- grid.415954.80000 0004 1771 3349Department of Radiology, China-Japan Friendship Hospital, Beijing, 100029 China
| | - Huaping Dai
- National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, 100029, Beijing, China. .,Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Chen Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, 130021, China. .,National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, 100029, Beijing, China. .,Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
15
|
Pulmonary Fibrosis as a Result of Acute Lung Inflammation: Molecular Mechanisms, Relevant In Vivo Models, Prognostic and Therapeutic Approaches. Int J Mol Sci 2022; 23:ijms232314959. [PMID: 36499287 PMCID: PMC9735580 DOI: 10.3390/ijms232314959] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Pulmonary fibrosis is a chronic progressive lung disease that steadily leads to lung architecture disruption and respiratory failure. The development of pulmonary fibrosis is mostly the result of previous acute lung inflammation, caused by a wide variety of etiological factors, not resolved over time and causing the deposition of fibrotic tissue in the lungs. Despite a long history of study and good coverage of the problem in the scientific literature, the effective therapeutic approaches for pulmonary fibrosis treatment are currently lacking. Thus, the study of the molecular mechanisms underlying the transition from acute lung inflammation to pulmonary fibrosis, and the search for new molecular markers and promising therapeutic targets to prevent pulmonary fibrosis development, remain highly relevant tasks. This review focuses on the etiology, pathogenesis, morphological characteristics and outcomes of acute lung inflammation as a precursor of pulmonary fibrosis; the pathomorphological changes in the lungs during fibrosis development; the known molecular mechanisms and key players of the signaling pathways mediating acute lung inflammation and pulmonary fibrosis, as well as the characteristics of the most common in vivo models of these processes. Moreover, the prognostic markers of acute lung injury severity and pulmonary fibrosis development as well as approved and potential therapeutic approaches suppressing the transition from acute lung inflammation to fibrosis are discussed.
Collapse
|
16
|
Dutt N, Shishir S, Chauhan NK, Jalandra R, kuwal A, Garg P, Kumar D, Vishwajeet V, Chakraborti A, Deokar K, Asfahan S, Babu A, bajad P, Gupta N, Khurana A, Garg MK. Mortality and Its Predictors in COVID-19 Patients With Pre-existing Interstitial Lung Disease. Cureus 2022; 14:e27759. [PMID: 36106257 PMCID: PMC9448685 DOI: 10.7759/cureus.27759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2022] [Indexed: 11/05/2022] Open
Abstract
Background The data on the impact of coronavirus disease 2019 (COVID-19) on interstitial lung disease (ILD) is still limited. To the best of our knowledge, there has been no study from India to date to assess the impact of COVID-19 in patients with preexisting ILD. We undertook this study to assess the clinical outcome of ILD patients admitted to our hospital with COVID-19. Methods In this retrospective observational study, records of reverse transcription polymerase chain reaction (RT-PCR)-confirmed COVID-19 patients with preexisting ILD who were admitted to the hospital in the period from May 1, 2020, to April 30, 2021, were obtained from the hospital database. The clinical outcomes of the patients were recorded. Univariate analysis was performed to find relation between various predetermined risk factors for mortality and those with significant p values (p<0.05) were subjected to multiple logistic regression to determine independent risk factors. Results In our study of 28 patients, the overall mortality was 35.7%. On comparing the parameters associated with increased mortality, there was no effect of age, gender, comorbidities, type of ILD, CT thorax findings on diagnosis, use of corticosteroids and antifibrotics in the past, spirometric findings on mortality. On multivariate analysis, the significant parameters were interleukin 6 (IL-6), p=0.02, OR=1.020 (1.006-1.043) and D-dimer, p=0.04, OR=2.14 (5.55-1.14). Conclusion COVID-19 in patients with pre-existing ILD has a comparatively higher mortality. D-dimer and IL-6 are significant predictors of mortality in ILD patients infected with COVID-19.
Collapse
|
17
|
Shrivastava G, Valenzuela-Leon PC, Chagas AC, Kern O, Botello K, Zhang Y, Martin-Martin I, Oliveira MB, Tirloni L, Calvo E. Alboserpin, the Main Salivary Anticoagulant from the Disease Vector Aedes albopictus, Displays Anti-FXa-PAR Signaling In Vitro and In Vivo. Immunohorizons 2022; 6:373-383. [PMID: 35738824 PMCID: PMC10753553 DOI: 10.4049/immunohorizons.2200045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/19/2022] Open
Abstract
Blood-feeding arthropods secrete potent salivary molecules, which include platelet aggregation inhibitors, vasodilators, and anticoagulants. Among these molecules, Alboserpin, the major salivary anticoagulant from the mosquito vector Aedes albopictus, is a specific inhibitor of the human coagulation factor Xa (FXa). In this study, we investigated the anti-inflammatory properties of Alboserpin, in vitro and in vivo. In vitro, Alboserpin inhibited FXa-induced protease-activated receptor (PAR)-1, PAR-2, PAR-3, VCAM, ICAM, and NF-κB gene expression in primary dermal microvascular endothelial cells. Alboserpin also prevented FXa-stimulated ERK1/2 gene expression and subsequent inflammatory cytokine release (MCP-1, TNF-α, IL-6, IL-8, IL-1β, IL-18). In vivo, Alboserpin reduced paw edema induced by FXa and subsequent release of inflammatory cytokines (CCL2, MCP-1, IL-1α, IL-6, IL-1β). Alboserpin also reduced FXa-induced endothelial permeability in vitro and in vivo. These findings show that Alboserpin is a potent anti-inflammatory molecule, in vivo and in vitro, and may play a significant role in blood feeding.
Collapse
Affiliation(s)
- Gaurav Shrivastava
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Paola Carolina Valenzuela-Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Andrezza Campos Chagas
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Olivia Kern
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Karina Botello
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Yixiang Zhang
- Protein Chemistry Section, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT; and
| | - Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Markus Berger Oliveira
- Tick-Pathogen Transmission Unit, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, Hamilton, MT
| | - Lucas Tirloni
- Tick-Pathogen Transmission Unit, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, Hamilton, MT
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD;
| |
Collapse
|
18
|
Oh H, Park HE, Song MS, Kim H, Baek JH. The Therapeutic Potential of Anticoagulation in Organ Fibrosis. Front Med (Lausanne) 2022; 9:866746. [PMID: 35652066 PMCID: PMC9148959 DOI: 10.3389/fmed.2022.866746] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/13/2022] [Indexed: 11/23/2022] Open
Abstract
Fibrosis, also known as organ scarring, describes a pathological stiffening of organs or tissues caused by increased synthesis of extracellular matrix (ECM) components. In the past decades, mounting evidence has accumulated showing that the coagulation cascade is directly associated with fibrotic development. Recent findings suggest that, under inflammatory conditions, various cell types (e.g., immune cells) participate in the coagulation process causing pathological outcomes, including fibrosis. These findings highlighted the potential of anticoagulation therapy as a strategy in organ fibrosis. Indeed, preclinical and clinical studies demonstrated that the inhibition of blood coagulation is a potential intervention for the treatment of fibrosis across all major organs (e.g., lung, liver, heart, and kidney). In this review, we aim to summarize our current knowledge on the impact of components of coagulation cascade on fibrosis of various organs and provide an update on the current development of anticoagulation therapy for fibrosis.
Collapse
|
19
|
Korfei M, Mahavadi P, Guenther A. Targeting Histone Deacetylases in Idiopathic Pulmonary Fibrosis: A Future Therapeutic Option. Cells 2022; 11:1626. [PMID: 35626663 PMCID: PMC9139813 DOI: 10.3390/cells11101626] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 02/07/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease with limited therapeutic options, and there is a huge unmet need for new therapies. A growing body of evidence suggests that the histone deacetylase (HDAC) family of transcriptional corepressors has emerged as crucial mediators of IPF pathogenesis. HDACs deacetylate histones and result in chromatin condensation and epigenetic repression of gene transcription. HDACs also catalyse the deacetylation of many non-histone proteins, including transcription factors, thus also leading to changes in the transcriptome and cellular signalling. Increased HDAC expression is associated with cell proliferation, cell growth and anti-apoptosis and is, thus, a salient feature of many cancers. In IPF, induction and abnormal upregulation of Class I and Class II HDAC enzymes in myofibroblast foci, as well as aberrant bronchiolar epithelium, is an eminent observation, whereas type-II alveolar epithelial cells (AECII) of IPF lungs indicate a significant depletion of many HDACs. We thus suggest that the significant imbalance of HDAC activity in IPF lungs, with a "cancer-like" increase in fibroblastic and bronchial cells versus a lack in AECII, promotes and perpetuates fibrosis. This review focuses on the mechanisms by which Class I and Class II HDACs mediate fibrogenesis and on the mechanisms by which various HDAC inhibitors reverse the deregulated epigenetic responses in IPF, supporting HDAC inhibition as promising IPF therapy.
Collapse
Affiliation(s)
- Martina Korfei
- Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, D-35392 Giessen, Germany; (P.M.); (A.G.)
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), D-35392 Giessen, Germany
| | - Poornima Mahavadi
- Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, D-35392 Giessen, Germany; (P.M.); (A.G.)
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), D-35392 Giessen, Germany
| | - Andreas Guenther
- Biomedical Research Center Seltersberg (BFS), Justus Liebig University Giessen, D-35392 Giessen, Germany; (P.M.); (A.G.)
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), D-35392 Giessen, Germany
- Lung Clinic, Evangelisches Krankenhaus Mittelhessen, D-35398 Giessen, Germany
- European IPF Registry and Biobank, D-35392 Giessen, Germany
| |
Collapse
|
20
|
Francis LRA, Millington-Burgess SL, Rahman T, Harper MT. Q94 is not a selective modulator of proteinase-activated receptor 1 (PAR1) in platelets. Platelets 2022; 33:1090-1095. [PMID: 35417662 DOI: 10.1080/09537104.2022.2026911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Thrombin is a potent platelet activator, acting through proteinase-activated receptors -1 and -4 (PAR1 and PAR4). Of these, PAR-1 is activated more rapidly and by lower thrombin concentrations. Consequently, PAR-1 has been extensively investigated as a target for anti-platelet drugs to prevent myocardial infarction. Q94 has been reported to act as an allosteric modulator of PAR1, potently and selectively inhibiting PAR1-Gαq coupling in multiple cell lines, but its effects on human platelet activation have not been previously studied. Platelet Ca2+ signaling, integrin αIIbβ3 activation and α-granule secretion were monitored following stimulation by a PAR1-activating peptide (PAR1-AP). Although Q94 inhibited these responses, its potency was low compared to other PAR1 antagonists. In addition, αIIbβ3 activation and α-granule secretion in response to other platelet activators were also inhibited with similar potency. Finally, in endothelial cells, Q94 did not inhibit PAR1-dependent Ca2+ signaling. Our data suggest that Q94 may have PAR1-independent off-target effects in platelets, precluding its use as a selective PAR1 allosteric modulator.
Collapse
Affiliation(s)
- Luc R A Francis
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
21
|
Zhang X, Chu C, Huang Y. Inhibition of thioredoxin-interacting protein may enhance the therapeutic effect of dehydrocostus lactone in cardiomyocytes under doxorubicin stimulation via the inhibition of the inflammatory response. Exp Ther Med 2022; 23:226. [PMID: 35222703 PMCID: PMC8812107 DOI: 10.3892/etm.2022.11150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/10/2021] [Indexed: 11/06/2022] Open
Abstract
Heart failure (HF) is the leading cause of death around the world, the mortality caused by HF is growing rapidly, and has become a great threaten to both public health and economic growth. Dehydrocostus lactone (DHE) is the active constituent of Saussurea lappa and is widely used in traditional Chinese medicine for its multiple biological functions, including anti-inflammatory, antioxidant and anti-cancer. To the best of our knowledge, DHE's effect on HF has not been clarified. Thioredoxin-interacting protein (TXNIP) regulates the process of oxidative stress and inflammation and leads to an increase in oxidative stress via oxidization of thioredoxin, TXNIP promotes the activation of the immune response by its binding with the NOD-like receptor protein 3 inflammasome. An MTT assay revealed that the overexpression or inhibition of TXNIP markedly decreased or significantly increased the proliferation of H9c2 cells, respectively. Through reverse transcription-quantitative PCR (RT-qPCR) and western blotting, it was determined that the expression of proinflammatory cytokines was significantly decreased with the increased expression of anti-inflammatory cytokines in a TXNIP knockout model. Further study utilizing RT-qPCR and western blotting demonstrated that these effects may be mediated by the nuclear factor erythroid 2-related factor 2/heme oxygenase-1/NF-κB signaling pathway. In conclusion, TXNIP inhibition may promote the therapeutic effect of DHE on oxidative stress-induced damage.
Collapse
Affiliation(s)
- Xuezhi Zhang
- Department of Critical Care Medicine, Anqiu People's Hospital, Anqiu, Shandong 262100, P.R. China
| | - Cuiyu Chu
- Department of Critical Care Medicine, Anqiu People's Hospital, Anqiu, Shandong 262100, P.R. China
| | - Yuankun Huang
- Department of Critical Care Medicine, Anqiu People's Hospital, Anqiu, Shandong 262100, P.R. China
| |
Collapse
|
22
|
Jaffar J, McMillan L, Wilson N, Panousis C, Hardy C, Cho HJ, Symons K, Glaspole I, Westall G, Wong M. Coagulation Factor-XII induces interleukin-6 by primary lung fibroblasts: A role in idiopathic pulmonary fibrosis? Am J Physiol Lung Cell Mol Physiol 2021; 322:L258-L272. [PMID: 34873957 DOI: 10.1152/ajplung.00165.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background The mechanisms driving idiopathic pulmonary fibrosis (IPF) remain undefined, however it is postulated that coagulation imbalances may play a role. The impact of blood-derived clotting factors, including factor XII (FXII) has not been investigated in the context of IPF. Methods Plasma levels of FXII were measured by ELISA in patients with IPF and age-matched healthy donors. Expression of FXII in human lung tissue was quantified using multiplex immunohistochemistry and western blotting. Mechanistic investigation of FXII activity was assessed in vitro on primary lung fibroblasts using qPCR and specific receptor/FXII inhibition. The functional outcome of FXII on fibroblast migration was examined by high-content image analysis. Findings Compared to 35 healthy donors, plasma levels of FXII were not higher in IPF (n=27, p>0·05). Tissue FXII was elevated in IPF (n=11) and increased numbers of FXII+ cells were found in IPF (n=8) lung tissue compared to non-diseased controls (n=6, p<0·0001). Activated FXII induced IL6 mRNA and IL-6 protein in fibroblasts that was blocked by anti-FXII antibody, CSL312. FXII-induced IL-6 production via PAR-1 and NF-kB. FXII induced migration of fibroblasts in a concentration-dependent manner. Interpretation FXII is normally confined to the circulation but leaks from damaged vessels into the lung interstitium in IPF where it 1) induces IL-6 production and 2) enhances migration of resident fibroblasts, critical events that drive chronic inflammation and therefore, contribute to fibrotic disease progression. Targeting FXII-induced fibroblastic processes in IPF may ameliorate pulmonary fibrosis. Funding National Health and Medical Research Council CRE in Lung Fibrosis and CSL Ltd.
Collapse
Affiliation(s)
- Jade Jaffar
- Department of Immunology and Pathology, Monash University, Australia.,Department of Respiratory Medicine, The Alfred Hospital, Australia
| | | | | | | | | | - Hyun Jung Cho
- Biological Optical Microscopy Platform, The University of Melbourne, Australia
| | - Karen Symons
- Department of Respiratory Medicine, The Alfred Hospital, Australia
| | - Ian Glaspole
- Department of Immunology and Pathology, Monash University, Australia.,Department of Respiratory Medicine, The Alfred Hospital, Australia
| | - Glen Westall
- Department of Immunology and Pathology, Monash University, Australia.,Department of Respiratory Medicine, The Alfred Hospital, Australia
| | - Mae Wong
- CSL Limited, Parkville, Victoria, Australia
| |
Collapse
|
23
|
Serafini FL, Lanuti P, Delli Pizzi A, Procaccini L, Villani M, Taraschi AL, Pascucci L, Mincuzzi E, Izzi J, Chiacchiaretta P, Buca D, Catitti G, Bologna G, Simeone P, Pieragostino D, Caulo M. Diagnostic Impact of Radiological Findings and Extracellular Vesicles: Are We Close to Radiovesicolomics? BIOLOGY 2021; 10:biology10121265. [PMID: 34943180 PMCID: PMC8698452 DOI: 10.3390/biology10121265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 11/23/2022]
Abstract
Simple Summary Over the years, diagnostic tests such as in radiology and flow cytometry have become more and more powerful in the constant struggle against different pathologies, some of which are life-threatening. The possibility of using these “weapons” in a conjugated manner could result in higher healing and prevention rates, and a decrease in late diagnosis diseases. Different correlations among pathologies, extracellular vesicles (EVs), and radiological findings were recently demonstrated by many authors. Together with the increasing importance of “omics” sciences, and artificial intelligence in this new century, the perspective of a new research field called “radiovesicolomics” could be the missing link, enabling a different approach to disease diagnosis and treatment. Abstract Currently, several pathologies have corresponding and specific diagnostic and therapeutic branches of interest focused on early and correct detection, as well as the best therapeutic approach. Radiology never ceases to develop newer technologies in order to give patients a clear, safe, early, and precise diagnosis; furthermore, in the last few years diagnostic imaging panoramas have been extended to the field of artificial intelligence (AI) and machine learning. On the other hand, clinical and laboratory tests, like flow cytometry and the techniques found in the “omics” sciences, aim to detect microscopic elements, like extracellular vesicles, with the highest specificity and sensibility for disease detection. If these scientific branches started to cooperate, playing a conjugated role in pathology diagnosis, what could be the results? Our review seeks to give a quick overview of recent state of the art research which investigates correlations between extracellular vesicles and the known radiological features useful for diagnosis.
Collapse
Affiliation(s)
- Francesco Lorenzo Serafini
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Andrea Delli Pizzi
- Institute of Advanced Biomedical Technologies (ITAB), University “G. d’Annunzio”, 66100 Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio”, 66100 Chieti, Italy
- Correspondence:
| | - Luca Procaccini
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Michela Villani
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Alessio Lino Taraschi
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Luca Pascucci
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Erica Mincuzzi
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Jacopo Izzi
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Piero Chiacchiaretta
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
- Institute of Advanced Biomedical Technologies (ITAB), University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Davide Buca
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Giulia Catitti
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Giuseppina Bologna
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Pasquale Simeone
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Damiana Pieragostino
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Massimo Caulo
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
- Institute of Advanced Biomedical Technologies (ITAB), University “G. d’Annunzio”, 66100 Chieti, Italy
| |
Collapse
|
24
|
Suppression of the fibrotic encapsulation of silicone implants by inhibiting the mechanical activation of pro-fibrotic TGF-β. Nat Biomed Eng 2021; 5:1437-1456. [PMID: 34031559 DOI: 10.1038/s41551-021-00722-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 04/07/2021] [Indexed: 02/07/2023]
Abstract
The fibrotic encapsulation of implants involves the mechanical activation of myofibroblasts and of pro-fibrotic transforming growth factor beta 1 (TGF-β1). Here, we show that both softening of the implant surfaces and inhibition of the activation of TGF-β1 reduce the fibrotic encapsulation of subcutaneous silicone implants in mice. Conventionally stiff silicones (elastic modulus, ~2 MPa) coated with a soft silicone layer (elastic modulus, ~2 kPa) reduced collagen deposition as well as myofibroblast activation without affecting the numbers of macrophages and their polarization states. Instead, fibroblasts around stiff implants exhibited enhanced intracellular stress, increased the recruitment of αv and β1 integrins, and activated TGF-β1 signalling. In vitro, the recruitment of αv integrin to focal adhesions and the activation of β1 integrin and of TGF-β were higher in myofibroblasts grown on latency-associated peptide (LAP)-coated stiff silicones than on soft silicones. Antagonizing αv integrin binding to LAP through the small-molecule inhibitor CWHM-12 suppressed active TGF-β signalling, myofibroblast activation and the fibrotic encapsulation of stiff subcutaneous implants in mice.
Collapse
|
25
|
Animal models of drug-induced pulmonary fibrosis: an overview of molecular mechanisms and characteristics. Cell Biol Toxicol 2021; 38:699-723. [PMID: 34741237 DOI: 10.1007/s10565-021-09676-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/21/2021] [Indexed: 01/08/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease characterized by progressive loss of pulmonary function. Drug-induced interstitial lung disease has been reported as a severe adverse effect of some drugs, such as bleomycin, amiodarone, and methotrexate. Based on good characteristics, drug-induced pulmonary fibrosis (PF) animal model has played a key role in our understanding of the molecular mechanisms of PF pathogenesis and recapitulates the specific pathology in patients and helps develop therapeutic strategies. Here, we summarize the mechanisms and characteristics of given fibrotic drug-induced animal models for PFs. Together with the key publications describing these models, this brief but detailed overview would be helpful for the pharmacological research with animal models of PFs. Potential mechanisms underlying drug induced lung toxicity.
Collapse
|
26
|
Han H, Zhao X, Liao M, Song Y, You C, Dong X, Yang X, Wang X, Huang B, Du M, Yan H. Activated Blood Coagulation Factor X (FXa) Contributes to the Development of Traumatic PVR Through Promoting RPE Epithelial-Mesenchymal Transition. Invest Ophthalmol Vis Sci 2021; 62:29. [PMID: 34283209 PMCID: PMC8300057 DOI: 10.1167/iovs.62.9.29] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Purpose Uncontrolled coagulation reactions contribute to pathological fibroproliferation in several organs, and yet their role in proliferative vitreoretinopathy (PVR) remains to be elucidated. In this study, we evaluated the profibrotic effects of FXa in RPE cells and in a mouse model of PVR. Methods FXa levels in the eyes of traumatic PVR patients and rabbit models of mechanical ocular trauma was measured by ELISA and immunohistochemistry. FXa-induced RPE EMT was assessed by examining cell proliferation, migration, tight junction changes, and expression of fibrotic markers. For in vivo study, FXa was injected into dispase-injured eyes, then intraocular fibrosis was evaluated by histological analysis and Western blotting. The therapeutic effect of FXa inhibitor was also examined in PVR mouse models. Results Vitreous FXa were higher in patients with traumatic PVR compared to patients with macular hole. Moreover, expressions of FXa and PAR1 were found in the epiretinal membranes from traumatic PVR patients. Vitreous FXa were markedly increased after mechanical ocular trauma in rabbits. In vitro, FXa stimulated RPE EMT characterized as ZO-1 disruption, compromised cell polarity, and increased fibronectin expressions. Co-injection of FXa and dispase in mice induced more severely damaged retinal structures, and increased α-SMA expressions than FXa or dispase treatment alone. Oral FXa or thrombin inhibitors significantly blocked intraocular fibrosis in PVR mouse models. FXa promoted phospho-activation of p38 in ARPE19 cells, which was dependent on PAR1. Moreover, TGF-βR inhibitor also significantly alleviated FXa-induced intraocular fibrosis in mice. Conclusions FXa promotes intraocular fibrosis in mice via mechanisms involving RPE activation.
Collapse
Affiliation(s)
- Han Han
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiao Zhao
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Mengyu Liao
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yinting Song
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Caiyun You
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xue Dong
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Molecular Ophthalmology, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xueli Yang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China.,Department of Ophthalmology, The First Affiliated Hospital of Dali University, Yunnan, China
| | - Xiaohong Wang
- Laboratory of Molecular Ophthalmology, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Bo Huang
- Department of Ophthalmology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Mei Du
- Laboratory of Molecular Ophthalmology, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
27
|
Risk of recurrent venous thromboembolism and bleeding in patients with interstitial lung disease: a cohort study. J Thromb Thrombolysis 2021; 53:67-73. [PMID: 34232453 DOI: 10.1007/s11239-021-02518-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 10/20/2022]
Abstract
Interstitial lung disease (ILD) encompasses various parenchymal lung disorders, which has the potential to increase the risk of venous thromboembolism (VTE). To evaluate, in patients with ILD and VTE, the risk of recurrent VTE during follow-up after stopping anticoagulation. This was a cohort of patients with a first VTE recruited between 1997 and 2015. The primary outcome was adjudicated fatal or nonfatal recurrent VTE after stopping anticoagulation. Main secondary outcomes were major or clinically relevant non-major bleeding under anticoagulation. Among 4314 patients with VTE, 50 had ILD diagnosed before VTE. Of these, anticoagulation was stopped in 30 patients after a median duration of 180 days and continued indefinitely in 20 patients. During a median follow-up of 27.8 months after anticoagulation discontinuation, recurrent VTE occurred in 15 on 30 patients (annual incidence of 19.2 events per 100-person-years [95%CI 12.0-29.3], case-fatality rate of 6.7% [95%CI 1.21-29.8]). The risk of recurrence was threefold higher when VTE was unprovoked and case-fatality rate of recurrence was increased by 3 when VTE index was PE. During the anticoagulant period, (median duration of 8.6 months), 6 patients had a major or clinically relevant bleeding (annual incidence of 7.3 events per 100-person-years [95%CI 3.4-15.1], case-fatality rate of 16.7% [95%CI 3.0-56.4]). In patients with ILD, the risk of recurrent VTE after stopping anticoagulation and the risk of bleeding under anticoagulation were very high. Our results suggest that anticoagulation should not be prolonged beyond 3-6 months of anticoagulation in most of cases.
Collapse
|
28
|
Chan SMH, Bernardo I, Mastronardo C, Mou K, De Luca SN, Seow HJ, Dobric A, Brassington K, Selemidis S, Bozinovski S, Vlahos R. Apocynin prevents cigarette smoking-induced loss of skeletal muscle mass and function in mice by preserving proteostatic signalling. Br J Pharmacol 2021; 178:3049-3066. [PMID: 33817783 PMCID: PMC8362135 DOI: 10.1111/bph.15482] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/08/2021] [Accepted: 03/24/2021] [Indexed: 12/15/2022] Open
Abstract
Background and Purpose Skeletal muscle dysfunction is a major comorbidity of chronic obstructive pulmonary disease (COPD). This type of muscle dysfunction may be a direct consequence of oxidative insults evoked by cigarette smoke (CS) exposure. The present study examined the effects of a potent Nox inhibitor and reactive oxygen species (ROS) scavenger, apocynin, on CS‐induced muscle dysfunction. Experimental Approach Male BALB/c mice were exposed to either room air (sham) or CS generated from nine cigarettes per day, 5 days a week for 8 weeks, with or without the coadministration of apocynin (5 mg·kg−1, i.p.). C2C12 myotubes exposed to either hydrogen peroxide (H2O2) or water‐soluble cigarette smoke extract (CSE) with or without apocynin (500 nM) were used as an experimental model in vitro. Key Results Eight weeks of CS exposure caused muscle dysfunction in mice, reflected by 10% loss of muscle mass and 54% loss of strength of tibialis anterior which were prevented by apocynin administration. In C2C12 myotubes, direct exposure to H2O2 or CSE caused myofibre wasting, accompanied by ~50% loss of muscle‐derived insulin‐like growth factor (IGF)‐1 and two‐fold induction of Cybb, independent of cellular inflammation. Expression of myostatin and MAFbx, negative regulators of muscle mass, were up‐regulated under H2O2 but not CSE conditions. Apocynin treatment abolished CSE‐induced Cybb expression, preserving muscle‐derived IGF‐1 expression and signalling pathway downstream of mammalian target of rapamycin (mTOR), thereby preventing myofibre wasting. Conclusion and Implications Targeted pharmacological inhibition of Nox‐derived ROS may alleviate the lung and systemic manifestations in smokers with COPD.
Collapse
Affiliation(s)
- Stanley M H Chan
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Ivan Bernardo
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Chanelle Mastronardo
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Kevin Mou
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Simone N De Luca
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Huei Jiunn Seow
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Aleksandar Dobric
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Kurt Brassington
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Ross Vlahos
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
29
|
Sun C, Ma Q, Yin J, Zhang H, Liu X. WISP-1 induced by mechanical stress contributes to fibrosis and hypertrophy of the ligamentum flavum through Hedgehog-Gli1 signaling. Exp Mol Med 2021; 53:1068-1079. [PMID: 34158608 PMCID: PMC8257797 DOI: 10.1038/s12276-021-00636-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/10/2021] [Accepted: 04/09/2021] [Indexed: 02/06/2023] Open
Abstract
Ongoing chronic fibrosis and hypertrophy of the ligamentum flavum (LF) is an important cause of lumbar spinal canal stenosis (LSCS). Our previous work showed that WNT1-inducible signaling pathway protein 1 (WISP-1) is a critical driver of LF fibrosis. However, the potential mechanism has not been explored. Here, we found that Gli1 was upregulated in hypertrophic LF tissues and required for fibrogenesis in fibroblasts. Moreover, mechanical stretching increased the expression of WISP-1 in LF fibroblasts. Furthermore, WISP-1 induced fibrogenesis in vitro through the Hedgehog-Gli1 pathway. This conclusion was supported by the fact that WISP-1 activated the Hedgehog-Gli1 pathway in LF fibroblasts and that cyclopamine attenuated the effect of WISP-1-induced fibrogenesis. WISP-1 also promoted the transition of fibroblasts into myofibroblasts via the Hedgehog pathway. Importantly, a hypertrophic LF rabbit model induced by mechanical stress also showed pathological changes in fibrosis and elevated expression of WISP-1, Gli1, and α-SMA. Therapeutic administration of cyclopamine reduced collagen expression, fibroblast proliferation, and myofibroblast differentiation and ameliorated fibrosis in the mechanical stress-induced rabbit model. Collectively, our findings show mechanical stress/WISP-1/Hedgehog signaling as a new fibrotic axis contributing to LF hypertrophy and identify Hedgehog signaling as a therapeutic target for the prevention and treatment of LF fibrosis.
Collapse
Affiliation(s)
- Chao Sun
- Department of Spine Surgery, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Qinghong Ma
- Department of Spine Surgery, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Jian Yin
- Department of Spine Surgery, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Han Zhang
- Department of Spine Surgery, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Xinhui Liu
- Department of Spine Surgery, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China.
| |
Collapse
|
30
|
Abstract
Myofibroblasts are critical to processes involved in normal wound healing and during pathological fibrosis. They transdifferentiate from fibroblasts, and in doing so become contractile and capable of secreting large amounts of extracellular matrix proteins. Transforming growth factor-beta (TGFβ) is a key cytokine involved in wound healing and fibrogenesis. TGFβ signaling has long been the subject of experimental therapeutic approaches to inhibit fibrosis in a variety of organ systems. Inhibition of TGFβ can reduce myofibroblast transdifferentiation, contractility, and matrix production. Importantly, TGFβ is released from cells and sequestered in the extracellular matrix in a latent form that requires activation for biological function. There have been multiple mechanisms of TGFβ activation described in a variety of cell types and in cell free systems; however, myofibroblasts have previously been shown to activate TGFβ via cell surface integrins, particularly αvβ5 integrins. This chapter will provide detailed protocols for accurately measuring activation of TGFβ by myofibroblasts in vitro. Levels of active TGFβ usually represent a small proportion of the total amount of latent TGFβ present in the matrix. Methods to measure active TGFβ therefore need to be sensitive and specific to detect the active cytokine only.
Collapse
Affiliation(s)
- Joanne Porte
- Division of Respiratory Medicine, NIHR Nottingham Respiratory Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Gisli Jenkins
- Division of Respiratory Medicine, NIHR Nottingham Respiratory Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Amanda L Tatler
- Division of Respiratory Medicine, NIHR Nottingham Respiratory Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK.
| |
Collapse
|
31
|
Guillotin D, Taylor AR, Platé M, Mercer PF, Edwards LM, Haggart R, Miele G, McAnulty RJ, Maher TM, Hynds RE, Jamal-Hanjani M, Marshall RP, Fisher AJ, Blanchard AD, Chambers RC. Transcriptome analysis of IPF fibroblastic foci identifies key pathways involved in fibrogenesis. Thorax 2021; 76:73-82. [PMID: 33214245 DOI: 10.1136/thoraxjnl-2020-214902] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Fibroblastic foci represent the cardinal pathogenic lesion in idiopathic pulmonary fibrosis (IPF) and comprise activated fibroblasts and myofibroblasts, the key effector cells responsible for dysregulated extracellular matrix deposition in multiple fibrotic conditions. The aim of this study was to define the major transcriptional programmes involved in fibrogenesis in IPF by profiling unmanipulated myofibroblasts within fibrotic foci in situ by laser capture microdissection. METHODS The challenges associated with deriving gene calls from low amounts of RNA and the absence of a meaningful comparator cell type were overcome by adopting novel data mining strategies and by using weighted gene co-expression network analysis (WGCNA), as well as an eigengene-based approach to identify transcriptional signatures, which correlate with fibrillar collagen gene expression. RESULTS WGCNA identified prominent clusters of genes associated with cell cycle, inflammation/differentiation, translation and cytoskeleton/cell adhesion. Collagen eigengene analysis revealed that transforming growth factor β1 (TGF-β1), RhoA kinase and the TSC2/RHEB axis formed major signalling clusters associated with collagen gene expression. Functional studies using CRISPR-Cas9 gene-edited cells demonstrated a key role for the TSC2/RHEB axis in regulating TGF-β1-induced mechanistic target of rapamycin complex 1 activation and collagen I deposition in mesenchymal cells reflecting IPF and other disease settings, including cancer-associated fibroblasts. CONCLUSION These data provide strong support for the human tissue-based and bioinformatics approaches adopted to identify critical transcriptional nodes associated with the key pathogenic cell responsible for fibrogenesis in situ and further identify the TSC2/RHEB axis as a potential novel target for interfering with excessive matrix deposition in IPF and other fibrotic conditions.
Collapse
Affiliation(s)
- Delphine Guillotin
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London, UK
| | - Adam R Taylor
- Fibrosis Discovery Performance Unit, Respiratory Therapy Area, Medicines Research Centre, GlaxoSmithKline R&D, Stevenage, UK
| | - Manuela Platé
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London, UK
| | - Paul F Mercer
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London, UK
| | - Lindsay M Edwards
- Fibrosis Discovery Performance Unit, Respiratory Therapy Area, Medicines Research Centre, GlaxoSmithKline R&D, Stevenage, UK
| | | | | | - Robin J McAnulty
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London, UK
| | - Toby M Maher
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Robert E Hynds
- Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute, London, UK
| | - Richard P Marshall
- Fibrosis Discovery Performance Unit, Respiratory Therapy Area, Medicines Research Centre, GlaxoSmithKline R&D, Stevenage, UK
| | - Andrew J Fisher
- Newcastle Fibrosis Research Group, Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
- Institute of Transplantation, Newcastle Upon Tyne Hospitals, Newcastle Upon Tyne, UK
| | - Andy D Blanchard
- Fibrosis Discovery Performance Unit, Respiratory Therapy Area, Medicines Research Centre, GlaxoSmithKline R&D, Stevenage, UK
| | - Rachel C Chambers
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, London, UK
| |
Collapse
|
32
|
Del Turco S, Vianello A, Ragusa R, Caselli C, Basta G. COVID-19 and cardiovascular consequences: Is the endothelial dysfunction the hardest challenge? Thromb Res 2020; 196:143-151. [PMID: 32871306 PMCID: PMC7451195 DOI: 10.1016/j.thromres.2020.08.039] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/17/2020] [Accepted: 08/26/2020] [Indexed: 12/22/2022]
Abstract
A Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) has become a pandemic disease named Coronavirus Disease-19 (COVID-19) of epochal dimension. The clinical spectrum of COVID-19 is wide, ranging from asymptomatic forms to severe pneumonia, sepsis and multiple organ dysfunction syndromes resulting in poor outcomes. Among the various consequences of severe COVID-19, cardiovascular (CV) collapse appears the most serious and potentially lethal. On the other hand, pre-existent CV comorbidities are also associated with higher mortality. The most reliable hypothetical pathogenetic mechanism for CV complications and cardiac injury in severe COVID-19 patients appears to be a sustained endothelial dysfunction, caused by the interplay of inflammation and coagulation. In this review, we survey papers addressing issues related to severe COVID-19, characterized by enhanced lung microvascular loss, hypercytokinemia, hypoxemia and thrombosis. We discuss about how the virus-induced downregulation of the angiotensin converting enzyme-2 (ACE2) receptor, used to enter the host cell, could affect the renin-angiotensin system, attempting to clarify the doubts about the use of ACE inhibitors and Angiotensin-II receptor blockers in COVID-19 patients. Finally, we point out how the delicate and physiological homeostatic function of the endothelium, which turns into a disastrous battlefield of the complex interaction between "cytokine and coagulative storms", can be irreparably compromised and result in systemic inflammatory complications.
Collapse
Affiliation(s)
- Serena Del Turco
- Institute of Clinical Physiology, CNR, San Cataldo Research Area, Via Moruzzi, 1, 56124 Pisa, Italy.
| | - Annamaria Vianello
- Department of Information Engineering, Telemedicine Section, University of Pisa, Italy
| | - Rosetta Ragusa
- Institute of Clinical Physiology, CNR, San Cataldo Research Area, Via Moruzzi, 1, 56124 Pisa, Italy
| | - Chiara Caselli
- Institute of Clinical Physiology, CNR, San Cataldo Research Area, Via Moruzzi, 1, 56124 Pisa, Italy
| | - Giuseppina Basta
- Institute of Clinical Physiology, CNR, San Cataldo Research Area, Via Moruzzi, 1, 56124 Pisa, Italy.
| |
Collapse
|
33
|
Ritzenthaler JD, Zhang M, Torres-Gonzalez E, Roman J. The Integrin Inhibitor Cilengitide and Bleomycin-Induced Pulmonary Fibrosis : Cilengitide and Bleomycin-Induced Pulmonary Fibrosis. Lung 2020; 198:947-955. [PMID: 33146772 DOI: 10.1007/s00408-020-00400-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/18/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE Fibroproliferation and excess deposition of extracellular matrix (ECM) are the pathologic hallmarks of idiopathic pulmonary fibrosis (IPF), a chronic progressive disorder with high mortality and suboptimal treatment options. Although the etiologic mechanisms responsible for the development and progression of IPF remain unclear, cell-ECM interactions and growth factors are considered important. Cilengitide is a cyclic RGD pentapeptide with anti-angiogenic activity that targets αvβ3, αvβ5 and α5β1, integrins known to mediate cell-ECM interactions and activate the pro-fibrotic growth factor Transforming Growth Factor beta (TGF-β). METHODS Cilengitide was studied in vitro with the use of NIH/3T3 cells and primary lung fibroblasts, and in vivo in the well-characterized bleomycin-induced lung injury model. The extent of ECM deposition was determined by RT-PCR, Western blot, histologic analysis and hydroxyproline assay of lung tissue. Bronchoalveolar lavage analysis was used to determine cell counts. RESULTS Cilengitide treatment of cultured fibroblasts showed decreased adhesion to vitronectin and fibronectin, both integrin-dependent events. Cilengitide also inhibited TGF-β-induced fibronectin gene expression and reduced the accumulation of mRNAs and protein for fibronectin and collagen type I. Both preventive and treatment effects of daily injections of cilengitide (20 mg/kg) failed to inhibit the development of pulmonary fibrosis as determined by histological analysis (Ashcroft scoring), bronchoalveolar lavage (BAL) fluid cell counts, and hydroxyproline content. CONCLUSIONS Overall, our data suggest that, despite its in vitro activity in fibroblasts, daily injections of cilengitide (20 mg/kg) did not inhibit the development of or ameliorate bleomycin-induced pulmonary fibrosis in mice.
Collapse
Affiliation(s)
- Jeffrey D Ritzenthaler
- Department of Medicine, Division of Pulmonary, Allergy and Critical Medicine, Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University, Jefferson Alumni Hall, 381, Philadelphia, PA, 19107, USA.
| | - Michael Zhang
- Department of Pharmacology & Toxicology, University of Louisville Health Sciences Center, Louisville, KY, USA.,University of Minnesota Medical School, Minneapolis, MN, USA
| | - Edilson Torres-Gonzalez
- Department of Medicine, Division of Pulmonary, Allergy and Critical Medicine, Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University, Jefferson Alumni Hall, 381, Philadelphia, PA, 19107, USA
| | - Jesse Roman
- Department of Pharmacology & Toxicology, University of Louisville Health Sciences Center, Louisville, KY, USA.,Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Louisville Health Sciences Center, Louisville, KY, USA.,Department of Medicine, Division of Pulmonary, Allergy and Critical Medicine, Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University, Jefferson Alumni Hall, 381, Philadelphia, PA, 19107, USA.,Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
34
|
Zhou IY, Tanabe KK, Fuchs BC, Caravan P. Collagen-targeted molecular imaging in diffuse liver diseases. Abdom Radiol (NY) 2020; 45:3545-3556. [PMID: 32737546 DOI: 10.1007/s00261-020-02677-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 07/10/2020] [Accepted: 07/18/2020] [Indexed: 12/14/2022]
Abstract
Liver fibrosis is a common pathway shared by all progressive chronic liver diseases (CLD) regardless of the underlying etiologies. With liver biopsy being the gold standard in assessing fibrosis degree, there is a large unmet clinical need to develop non-invasive imaging tools that can directly and repeatedly quantify fibrosis throughout the liver for a more accurate assessment of disease burden, progression, and treatment response. Type I collagen is a particularly attractive target for molecular imaging as its excessive deposition is specific to fibrosis, and it is present in concentrations suitable for many imaging modalities. Novel molecular MRI contrast agents designed to bind with collagen provide direct quantification of collagen deposition, which have been validated across animal species and liver injury models. Collagen-targeted molecular imaging probes hold great promise not only as a tool for initial staging and surveillance of fibrosis progression, but also as a marker of fibrosis regression in drug trials.
Collapse
Affiliation(s)
- Iris Y Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Harvard Medical School, 149 13th St, Boston, MA, 02129, USA
- Institute for Innovation in Imaging (i3), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Kenneth K Tanabe
- Division of Surgical Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA.
- Harvard Medical School, 149 13th St, Boston, MA, 02129, USA.
- Institute for Innovation in Imaging (i3), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
35
|
Zhou IY, Catalano OA, Caravan P. Advances in functional and molecular MRI technologies in chronic liver diseases. J Hepatol 2020; 73:1241-1254. [PMID: 32585160 PMCID: PMC7572718 DOI: 10.1016/j.jhep.2020.06.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023]
Abstract
MRI has emerged as the most comprehensive non-invasive diagnostic tool for liver diseases. In recent years, the value of MRI in hepatology has been significantly enhanced by a wide range of contrast agents, both clinically available and under development, that add functional information to anatomically detailed morphological images, or increase the distinction between normal and pathological tissues by targeting molecular and cellular events. Several classes of contrast agents are available for contrast-enhanced hepatic MRI, including i) conventional non-specific extracellular fluid contrast agents for assessing tissue perfusion; ii) hepatobiliary-specific contrast agents that are taken up by functioning hepatocytes and excreted through the biliary system for evaluating hepatobiliary function; iii) superparamagnetic iron oxide particles that accumulate in Kupffer cells; and iv) novel molecular contrast agents that are biochemically targeted to specific molecular/cellular processes for staging liver diseases or detecting treatment responses. The use of different functional and molecular MRI methods enables the non-invasive assessment of disease burden, progression, and treatment response in a variety of liver diseases. A high diagnostic performance can be achieved with MRI by combining imaging biomarkers.
Collapse
Affiliation(s)
- Iris Y Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States; Harvard Medical School, Boston, MA, USA; Institute for Innovation in Imaging (i(3)), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Onofrio A Catalano
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States; Harvard Medical School, Boston, MA, USA; Division of Abdominal Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States; Harvard Medical School, Boston, MA, USA; Institute for Innovation in Imaging (i(3)), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
36
|
Stelcer E, Milecka P, Komarowska H, Jopek K, Tyczewska M, Szyszka M, Lesniczak M, Suchorska W, Bekova K, Szczepaniak B, Ruchala M, Karczewski M, Wierzbicki T, Szaflarski W, Malendowicz LK, Rucinski M. Adropin Stimulates Proliferation and Inhibits Adrenocortical Steroidogenesis in the Human Adrenal Carcinoma (HAC15) Cell Line. Front Endocrinol (Lausanne) 2020; 11:561370. [PMID: 33133015 PMCID: PMC7579427 DOI: 10.3389/fendo.2020.561370] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/04/2020] [Indexed: 12/21/2022] Open
Abstract
Adropin is a multifunctional peptide hormone encoded by the ENHO (energy homeostasis associated) gene. It plays a role in mechanisms related to increased adiposity, insulin resistance, as well as glucose, and lipid metabolism. The low adropin levels are strongly associated with obesity independent insulin resistance. On the other hand, overexpression or exogenous administration of adropin improves glucose homeostasis. The multidirectional, adropin-related effects associated with the regulation of metabolism in humans also appear to be attributable to the effects of this peptide on the activity of various elements of the endocrine system including adrenal cortex. Therefore, the main purpose of the present study was to investigate the effect of adropin on proliferation and secretory activity in the human HAC15 adrenal carcinoma cell line. In this study, we obtained several highly interesting findings. First, GPR19, the main candidate sensitizer of adrenocortical cells to adropin, was expressed in HAC15 cells. Moreover, GPR19 expression was relatively stable and not regulated by ACTH, forskolin, or adropin itself. Our findings also suggest that adropin has the capacity to decrease expression levels of steroidogenic genes such as steroidogenic acute regulatory protein (StAR) and CYP11A1, which then led to a statistically significant inhibition in cortisol and aldosterone biosynthesis and secretion. Based on whole transcriptome study and research involving transforming growth factor (TGF)-β type I receptor kinase inhibitor we demonstrated that attenuation of steroidogenesis caused by adropin is mediated by the TGF-β signaling pathway likely to act through transactivation mechanism. We found that HAC15 cells treated with adropin presented significantly higher proliferation levels than untreated cells. Using specific intracellular inhibitors, we showed that adropin stimulate proliferation via ERK1/2 and AKT dependent signaling pathways. We have also demonstrated that expression of GPR19 is elevated in adrenocortical carcinoma in relation to normal adrenal glands. High level of GPR19 expression in adrenocortical carcinoma may constitute a negative prognostic factor of disease progression.
Collapse
Affiliation(s)
- Ewelina Stelcer
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
- Radiobiology Lab, Greater Poland Cancer Centre, Poznan, Poland
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Paulina Milecka
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Hanna Komarowska
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Karol Jopek
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marianna Tyczewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marta Szyszka
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marta Lesniczak
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Wiktoria Suchorska
- Radiobiology Lab, Greater Poland Cancer Centre, Poznan, Poland
- Department of Electroradiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Karlygash Bekova
- West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Beata Szczepaniak
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marek Ruchala
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Marek Karczewski
- Department of General and Transplantation Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Tomasz Wierzbicki
- Department of General, Endocrinological and Gastroenterological Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Witold Szaflarski
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Ludwik K. Malendowicz
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marcin Rucinski
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
37
|
Frydman GH, Streiff MB, Connors JM, Piazza G. The Potential Role of Coagulation Factor Xa in the Pathophysiology of COVID-19: A Role for Anticoagulants as Multimodal Therapeutic Agents. ACTA ACUST UNITED AC 2020; 4:e288-e299. [PMID: 33043235 PMCID: PMC7541169 DOI: 10.1055/s-0040-1718415] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023]
Abstract
SARS-CoV-2 infection (COVID-19) results in local and systemic activation of inflammation and coagulation. In this review article, we will discuss the potential role of coagulation factor Xa (FXa) in the pathophysiology of COVID-19. FXa, a serine protease, has been shown to play a role in the cleavage of SARS-CoV-1 spike protein (SP), with the inhibition of FXa resulting in the inhibition of viral infectivity. FX is known to be primarily produced in the liver, but it is also expressed by multiple cells types, including alveolar epithelium, cardiac myocytes, and macrophages. Considering that patients with preexisting conditions, including cardiopulmonary disease, are at an increased risk of severe COVID-19, we discuss the potential role of increased levels of FX in these patients, resulting in a potential increased propensity to have a higher infectious rate and viral load, increased activation of coagulation and inflammation, and development of fibrosis. With these observations in mind, we postulate as to the potential therapeutic role of FXa inhibitors as a prophylactic and therapeutic treatment for high-risk patients with COVID-19.
Collapse
Affiliation(s)
- Galit H Frydman
- Coagulo Medical Technologies, Inc., Auburndale, Massachusetts, United States.,Center for Biomedical Engineering, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States.,Division of Trauma, Emergency Surgery and Surgical Critical Care, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Michael B Streiff
- Division of Hematology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Jean M Connors
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Gregory Piazza
- Division of Cardiovascular Medicine Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States
| |
Collapse
|
38
|
John AE, Graves RH, Pun KT, Vitulli G, Forty EJ, Mercer PF, Morrell JL, Barrett JW, Rogers RF, Hafeji M, Bibby LI, Gower E, Morrison VS, Man Y, Roper JA, Luckett JC, Borthwick LA, Barksby BS, Burgoyne RA, Barnes R, Le J, Flint DJ, Pyne S, Habgood A, Organ LA, Joseph C, Edwards-Pritchard RC, Maher TM, Fisher AJ, Gudmann NS, Leeming DJ, Chambers RC, Lukey PT, Marshall RP, Macdonald SJF, Jenkins RG, Slack RJ. Translational pharmacology of an inhaled small molecule αvβ6 integrin inhibitor for idiopathic pulmonary fibrosis. Nat Commun 2020; 11:4659. [PMID: 32938936 PMCID: PMC7494911 DOI: 10.1038/s41467-020-18397-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 08/17/2020] [Indexed: 12/16/2022] Open
Abstract
The αvβ6 integrin plays a key role in the activation of transforming growth factor-β (TGFβ), a pro-fibrotic mediator that is pivotal to the development of idiopathic pulmonary fibrosis (IPF). We identified a selective small molecule αvβ6 RGD-mimetic, GSK3008348, and profiled it in a range of disease relevant pre-clinical systems. To understand the relationship between target engagement and inhibition of fibrosis, we measured pharmacodynamic and disease-related end points. Here, we report, GSK3008348 binds to αvβ6 with high affinity in human IPF lung and reduces downstream pro-fibrotic TGFβ signaling to normal levels. In human lung epithelial cells, GSK3008348 induces rapid internalization and lysosomal degradation of the αvβ6 integrin. In the murine bleomycin-induced lung fibrosis model, GSK3008348 engages αvβ6, induces prolonged inhibition of TGFβ signaling and reduces lung collagen deposition and serum C3M, a marker of IPF disease progression. These studies highlight the potential of inhaled GSK3008348 as an anti-fibrotic therapy. The αvβ6 integrin is key in activating the pro-fibrotic cytokine TGFβ in idiopathic pulmonary fibrosis. Here, the authors show an inhaled small molecule αvβ6 inhibitor GSK3008348 induces prolonged inhibition of TGFβ signaling pathways in human and murine models of lung fibrosis via αvβ6 degradation.
Collapse
Affiliation(s)
- Alison E John
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Rebecca H Graves
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - K Tao Pun
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Giovanni Vitulli
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Ellen J Forty
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| | - Paul F Mercer
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| | - Josie L Morrell
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - John W Barrett
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Rebecca F Rogers
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Maryam Hafeji
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Lloyd I Bibby
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Elaine Gower
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Valerie S Morrison
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Yim Man
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - James A Roper
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Jeni C Luckett
- Radiological Sciences, University of Nottingham, Nottingham, UK
| | - Lee A Borthwick
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Ben S Barksby
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Rachel A Burgoyne
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Rory Barnes
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Joelle Le
- Drug Design and Selection - Molecular Design, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - David J Flint
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Anthony Habgood
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Louise A Organ
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Chitra Joseph
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | | | - Toby M Maher
- NIHR Respiratory Clinical Research Facility, Royal Brompton Hospital, London, UK.,Fibrosis Research Group, National Heart and Lung Institute, Imperial College, London, UK
| | - Andrew J Fisher
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK.,Institute of Transplantation, Freeman Hospital, Newcastle Upon Tyne Hospitals NHS, Foundation Trust, Newcastle upon Tyne, UK
| | - Natasja Stæhr Gudmann
- Nordic Bioscience A/S, Biomarkers and Research, Herlev Hovedgade 205-207, Herlev, Denmark
| | - Diana J Leeming
- Nordic Bioscience A/S, Biomarkers and Research, Herlev Hovedgade 205-207, Herlev, Denmark
| | - Rachel C Chambers
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| | - Pauline T Lukey
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Richard P Marshall
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Simon J F Macdonald
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - R Gisli Jenkins
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK.
| | - Robert J Slack
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| |
Collapse
|
39
|
Landi C, Bergantini L, Cameli P, d'Alessandro M, Carleo A, Shaba E, Rottoli P, Bini L, Bargagli E. Idiopathic Pulmonary Fibrosis Serum proteomic analysis before and after nintedanib therapy. Sci Rep 2020; 10:9378. [PMID: 32523095 PMCID: PMC7287088 DOI: 10.1038/s41598-020-66296-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 05/16/2020] [Indexed: 11/30/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal progressive disease with a median survival of 2–5 years. Nintedanib is a small tyrosine kinase inhibitor that reduces IPF progression, significantly slowing the annual decline in Forced Vital Capacity (FVC). Very little data is available on the molecular mechanisms of this treatment in IPF, despite a growing interest in the definition of IPF pathogenesis and target therapy. A functional proteomic approach was applied to the analysis of serum samples from IPF patients in order to highlight differential proteins potentially indicative of drug-induced molecular pathways modifications and response to therapy. Twelve serum samples were collected from six IPF patients in care at Siena Regional Referral Center for Interstitial Lung Diseases (ILDs) and treated with nintedanib for one year. Serum samples were analyzed at baseline (T0 before starting therapy) and after one year of treatment (T1) and underwent differential proteomic and bioinformatic analysis. Proteomic analysis revealed 13 protein species that were significantly increased after one year of treatment. When the targets of nintedanib (VEGFR, FGFR and PDGFR) were added, enrichment analysis extracted molecular pathways and process networks involved in cell differentiation (haptoglobin and albumin), coagulation (antithrombin III), epithelial mesenchymal transition, cell proliferation and transmigration. PI3K and MAPK induced up-regulation of apolipoprotein C3. Proteomic study found 13 protein species up-regulated in IPF patients after one year of nintedanib treatment. Haptoglobin, a central hub of our analysis was validated by 2D-WB and ELISA as theranostic marker in a more numerous populations of patients.
Collapse
Affiliation(s)
- Claudia Landi
- Functional Proteomics Lab, Dept. Life Sciences, University of Siena, Siena, Italy. .,Respiratory Diseases and Lung Transplant Unit, Dept. Internal and Specialist Medicine, AOUS, Siena, Italy.
| | - Laura Bergantini
- Respiratory Diseases and Lung Transplant Unit, Dept. Internal and Specialist Medicine, AOUS, Siena, Italy
| | - Paolo Cameli
- Respiratory Diseases and Lung Transplant Unit, Dept. Internal and Specialist Medicine, AOUS, Siena, Italy
| | - Miriana d'Alessandro
- Respiratory Diseases and Lung Transplant Unit, Dept. Internal and Specialist Medicine, AOUS, Siena, Italy
| | - Alfonso Carleo
- Department of Pneumology, Medical School Hannover (MHH), Hannover, Germany
| | - Enxhi Shaba
- Functional Proteomics Lab, Dept. Life Sciences, University of Siena, Siena, Italy
| | - Paola Rottoli
- Respiratory Diseases and Lung Transplant Unit, Dept. Internal and Specialist Medicine, AOUS, Siena, Italy
| | - Luca Bini
- Functional Proteomics Lab, Dept. Life Sciences, University of Siena, Siena, Italy
| | - Elena Bargagli
- Respiratory Diseases and Lung Transplant Unit, Dept. Internal and Specialist Medicine, AOUS, Siena, Italy
| |
Collapse
|
40
|
Kreuter M, Maher TM. Treatment of Acute Exacerbation of Idiopathic Pulmonary Fibrosis. A Call to Arms. Am J Respir Crit Care Med 2020; 201:1030-1032. [PMID: 31978310 PMCID: PMC7193842 DOI: 10.1164/rccm.202001-0057ed] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Michael Kreuter
- Center for Interstitial and Rare Lung Diseases, ThoraxklinikUniversity of HeidelbergHeidelberg, Germany
- German Center for Lung ResearchHeidelberg, Germany
| | - Toby M Maher
- National Institute for Health Research Respiratory Biomedical Research UnitRoyal Brompton HospitalLondon, United Kingdomand
- National Heart and Lung InstituteImperial CollegeLondon, United Kingdom
| |
Collapse
|
41
|
Guo X, Kolpakov MA, Hooshdaran B, Schappell W, Wang T, Eguchi S, Elliott KJ, Tilley DG, Rao AK, Andrade-Gordon P, Bunce M, Madhu C, Houser SR, Sabri A. Cardiac Expression of Factor X Mediates Cardiac Hypertrophy and Fibrosis in Pressure Overload. ACTA ACUST UNITED AC 2020; 5:69-83. [PMID: 32043021 PMCID: PMC7000872 DOI: 10.1016/j.jacbts.2019.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 10/04/2019] [Accepted: 10/07/2019] [Indexed: 11/28/2022]
Abstract
Factor X expression was increased in the heart following pressure overload and in isolated cardiac myocytes and fibroblasts. Rivaroxaban treatment at doses that do not affect thrombin generation, blood coagulation or cardiac hemostasis attenuated cardiac inflammation, hypertrophy, and fibrosis caused by pressure overload and improved cardiac diastolic function. Activated coagulation factor X induced PAR-1/-2–mediated elongated cardiomyocyte hypertrophy and PAR1-mediated cardiac fibroblast proliferation, migration and differentiation. Activated coagulation factor X derived from a cardiac source may represent an important physiologic and pathophysiologic activator of PAR-1/PAR-2. Non-anticoagulation dosage of rivaroxaban could provide an effective therapy to attenuate early phases of heart failure development.
Activated factor X is a key component of the coagulation cascade, but whether it directly regulates pathological cardiac remodeling is unclear. In mice subjected to pressure overload stress, cardiac factor X mRNA expression and activity increased concurrently with cardiac hypertrophy, fibrosis, inflammation and diastolic dysfunction, and responses blocked with a low coagulation-independent dose of rivaroxaban. In vitro, neurohormone stressors increased activated factor X expression in both cardiac myocytes and fibroblasts, resulting in activated factor X-mediated activation of protease-activated receptors and pro-hypertrophic and -fibrotic responses, respectively. Thus, inhibition of cardiac-expressed activated factor X could provide an effective therapy for the prevention of adverse cardiac remodeling in hypertensive patients.
Collapse
Affiliation(s)
- Xinji Guo
- Cardiovascular Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Mikhail A Kolpakov
- Cardiovascular Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Bahman Hooshdaran
- Cardiovascular Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - William Schappell
- Cardiovascular Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Tao Wang
- Cardiovascular Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Satoru Eguchi
- Cardiovascular Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Katherine J Elliott
- Cardiovascular Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Douglas G Tilley
- Center of Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - A Koneti Rao
- Sol Sherry Thrombosis Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | | | | | | | - Steven R Houser
- Cardiovascular Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Abdelkarim Sabri
- Cardiovascular Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
42
|
Fan C, Wang J, Mao C, Li W, Liu K, Wang Z. The FGL2 prothrombinase contributes to the pathological process of experimental pulmonary hypertension. J Appl Physiol (1985) 2019; 127:1677-1687. [PMID: 31580221 DOI: 10.1152/japplphysiol.00396.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In situ thrombus formation is one of the major pathological features of pulmonary hypertension (PH). The mechanism of in situ thrombosis has not been clearly identified. Fibrinogen-like protein 2 (FGL2) prothrombinase is an immune coagulant that can cleave prothrombin to thrombin, which then converts fibrinogen into fibrin. This mechanism triggers in situ thrombus formation directly, bypassing both the intrinsic and extrinsic coagulation pathways. FGL2 prothrombinase is mainly expressed in endothelial cells and mediates multiple pathological processes. This implies that it may also play a role in PH. In this study, we examined the expression of FGL2 in idiopathic pulmonary arterial hypertension (IPAH) patients, and in monocrotaline-induced rat and hypoxia-induced mouse PH models. Fgl2−/− mice were used to evaluate the development of PH and explore associated pathological changes. These included in situ thrombosis, vascular remodeling, and endothelial apoptosis. Following these analyses, we examined possible signaling pathways downstream of FGL2 in PH. We show FGL2 is upregulated in pulmonary vascular endothelium in human IPAH and in two animal PH models. Genetic knockout of Fgl2 limited the development of PH, indicated by decreased in situ thrombus formation, less vascular remodeling, and reduced endothelial dysfunction. In addition, loss of FGL2 downregulated PAR1 (proteinase-activated receptor 1) expression and decreased the overactivation and consumption of platelets in hypoxia-induced PH. These results indicate FGL2 participate in the development of PH and loss of FGL2 could attenuate PH by reducing in situ thrombosis and suppressing PAR1 signaling. Thus we provide evidence that suggests FGL2 prothrombinase presents a potential therapeutic target for clinical treatment of PH. NEW & NOTEWORTHY This is the first study to demonstrate that fibrinogen-like protein 2 (FGL2) participates in the pathological progression of pulmonary hypertension (PH) in human idiopathic pulmonary arterial hypertension, a monocrotaline rat PH model, and a hypoxia mouse PH model. Genetic knockout of Fgl2 significantly limited the development of PH indicated by reduced in situ thrombosis, vascular remodeling, and endothelial dysfunction, and suppressed PAR1 (proteinase-activated receptor 1) signaling and overactivation of platelets on PH. These results suggest FGL2 presents a potential therapeutic target for clinical treatment of PH.
Collapse
Affiliation(s)
- Cheng Fan
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jue Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoqin Mao
- Department of Rehabilitation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenzhu Li
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kun Liu
- Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaohui Wang
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Somogyi V, Chaudhuri N, Torrisi SE, Kahn N, Müller V, Kreuter M. The therapy of idiopathic pulmonary fibrosis: what is next? Eur Respir Rev 2019; 28:190021. [PMID: 31484664 PMCID: PMC9488691 DOI: 10.1183/16000617.0021-2019] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrosing interstitial lung disease, characterised by progressive scarring of the lung and associated with a high burden of disease and early death. The pathophysiological understanding, clinical diagnostics and therapy of IPF have significantly evolved in recent years. While the recent introduction of the two antifibrotic drugs pirfenidone and nintedanib led to a significant reduction in lung function decline, there is still no cure for IPF; thus, new therapeutic approaches are needed. Currently, several clinical phase I-III trials are focusing on novel therapeutic targets. Furthermore, new approaches in nonpharmacological treatments in palliative care, pulmonary rehabilitation, lung transplantation, management of comorbidities and acute exacerbations aim to improve symptom control and quality of life. Here we summarise new therapeutic attempts and potential future approaches to treat this devastating disease.
Collapse
Affiliation(s)
- Vivien Somogyi
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
- Dept of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Nazia Chaudhuri
- Manchester University NHS Foundation Trust, Wythenshawe Hospital, Manchester, UK
| | - Sebastiano Emanuele Torrisi
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
- Regional Referral Centre for Rare Lung Diseases, University Hospital "Policlinico", Dept of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Nicolas Kahn
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| | - Veronika Müller
- Dept of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Michael Kreuter
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
44
|
Clay TA, Steffen MA, Treglia ML, Torres CD, Trujano-Alvarez AL, Bonett RM. Multiple stressors produce differential transcriptomic patterns in a stream-dwelling salamander. BMC Genomics 2019; 20:482. [PMID: 31185901 PMCID: PMC6560913 DOI: 10.1186/s12864-019-5814-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 05/20/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Global biodiversity is decreasing at an alarming rate and amphibians are at the forefront of this crisis. Understanding the factors that negatively impact amphibian populations and effectively monitoring their health are fundamental to addressing this epidemic. Plasma glucocorticoids are often used to assess stress in amphibians and other vertebrates, but these hormones can be extremely dynamic and impractical to quantify in small organisms. Transcriptomic responses to stress hormones in amphibians have been largely limited to laboratory models, and there have been few studies on vertebrates that have evaluated the impact of multiple stressors on patterns of gene expression. Here we examined the gene expression patterns in tail tissues of stream-dwelling salamanders (Eurycea tynerensis) chronically exposed to the stress hormone corticosterone under different temperature regimes. RESULTS We found unique transcriptional signatures for chronic corticosterone exposure that were independent of temperature variation. Several of the corticosterone responsive genes are known to be involved in immune system response (LY-6E), oxidative stress (GSTM2 and TRX), and tissue repair (A2M and FX). We also found many genes to be influenced by temperature (CIRBP, HSC71, HSP40, HSP90, HSP70, ZNF593). Furthermore, the expression patterns of some genes (GSTM2, LY-6E, UMOD, ZNF593, CIRBP, HSP90) show interactive effects of temperature and corticosterone exposure, compared to each treatment alone. Through a series of experiments we also showed that stressor induced patterns of expression were largely consistent across ages, life cycle modes, and tissue regeneration. CONCLUSIONS Outside of thermal stressors, the application of transcriptomes to monitor the health of non-human vertebrate systems has been vastly underinvestigated. Our study suggests that transcriptomic patterns harbor stressor specific signatures that can be highly informative for monitoring the diverse stressors of amphibian populations.
Collapse
Affiliation(s)
- Timothy A Clay
- Department of Biological Science, University of Tulsa, Tulsa, OK, 74104, USA. .,Present Address: Department of Biological Sciences, Nicholls State University, Thibodaux, LA, 70310, USA.
| | - Michael A Steffen
- Department of Biological Science, University of Tulsa, Tulsa, OK, 74104, USA.,Present Address: Department of Biological Sciences, University of New Hampshire, Durham, NH, 03824, USA
| | - Michael L Treglia
- Department of Biological Science, University of Tulsa, Tulsa, OK, 74104, USA.,Present Address: The Nature Conservancy, New York, NY, 10001, USA
| | - Carolyn D Torres
- Department of Biological Science, University of Tulsa, Tulsa, OK, 74104, USA
| | | | - Ronald M Bonett
- Department of Biological Science, University of Tulsa, Tulsa, OK, 74104, USA.
| |
Collapse
|
45
|
van Cleemput J, Sonaglioni A, Wuyts WA, Bengus M, Stauffer JL, Harari S. Idiopathic Pulmonary Fibrosis for Cardiologists: Differential Diagnosis, Cardiovascular Comorbidities, and Patient Management. Adv Ther 2019; 36:298-317. [PMID: 30554332 PMCID: PMC6824347 DOI: 10.1007/s12325-018-0857-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Indexed: 02/06/2023]
Abstract
The presence of rare comorbidities in patients with cardiovascular disease (CVD) presents a diagnostic challenge to cardiologists. In evaluating these patients, cardiologists are faced with a unique opportunity to shorten diagnosis times and direct patients towards correct treatment pathways. Idiopathic pulmonary fibrosis (IPF), a type of interstitial lung disease (ILD), is an example of a rare disease where patients frequently demonstrate comorbid CVD. Both CVD and IPF most commonly affect a similar patient demographic: men over the age of 60 years with a history of smoking. Moreover, IPF and heart failure (HF) share a number of symptoms. As a result, patients with IPF can be misdiagnosed with HF and vice versa. This article aims to increase awareness of IPF among cardiologists, providing an overview for cardiologists on the differential diagnosis of IPF from HF, and describing the signs and symptoms that would warrant referral to a pulmonologist with expertise in ILD. Once patients with IPF have received a diagnosis, cardiologists can have an important role in managing patients who are candidates for a lung transplant or those who develop pulmonary hypertension (PH). Group 3 PH is one of the most common cardiovascular complications diagnosed in patients with IPF, its prevalence varying between reports but most often cited as between 30% and 50%. This review summarizes the current knowledge on Group 3 PH in IPF, discusses data from clinical trials assessing treatments for Group 1 PH in patients with IPF, and highlights that treatment guidelines recommend against these therapies in IPF. Finally, this article provides the cardiologist with an overview on the use of the two approved treatments for IPF, the antifibrotics pirfenidone and nintedanib, in patients with IPF and CVD comorbidities. Conversely, the impact of treatments for CVD comorbidities on patients with IPF is also discussed.Funding: F. Hoffmann-La Roche, Ltd.Plain Language Summary: Plain language summary available for this article.
Collapse
Affiliation(s)
| | - Andrea Sonaglioni
- U.O. di Cardiologia, Ospedale San Giuseppe - MultiMedica IRCCS, Milan, Italy
| | - Wim A Wuyts
- Department of Respiratory Medicine, Unit for Interstitial Lung Diseases, University Hospitals Leuven, Leuven, Belgium
| | | | | | - Sergio Harari
- U.O. di Pneumologia e Terapia Semi-Intensiva Respiratoria - Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, Ospedale San Giuseppe - MultiMedica IRCCS, Milan, Italy
| |
Collapse
|
46
|
Montesi SB, Désogère P, Fuchs BC, Caravan P. Molecular imaging of fibrosis: recent advances and future directions. J Clin Invest 2019; 129:24-33. [PMID: 30601139 DOI: 10.1172/jci122132] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Fibrosis, the progressive accumulation of connective tissue that occurs in response to injury, causes irreparable organ damage and may result in organ failure. The few available antifibrotic treatments modify the rate of fibrosis progression, but there are no available treatments to reverse established fibrosis. Thus, more effective therapies are urgently needed. Molecular imaging is a promising biomedical methodology that enables noninvasive visualization of cellular and subcellular processes. It provides a unique means to monitor and quantify dysregulated molecular fibrotic pathways in a noninvasive manner. Molecular imaging could be used for early detection, disease staging, and prognostication, as well as for assessing disease activity and treatment response. As fibrotic diseases are often molecularly heterogeneous, molecular imaging of a specific pathway could be used for patient stratification and cohort enrichment with the goal of improving clinical trial design and feasibility and increasing the ability to detect a definitive outcome for new therapies. Here we review currently available molecular imaging probes for detecting fibrosis and fibrogenesis, the active formation of new fibrous tissue, and their application to models of fibrosis across organ systems and fibrotic processes. We provide our opinion as to the potential roles of molecular imaging in human fibrotic diseases.
Collapse
Affiliation(s)
| | - Pauline Désogère
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Athinoula A. Martinos Center for Biomedical Imaging and.,Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bryan C Fuchs
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Caravan
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Athinoula A. Martinos Center for Biomedical Imaging and.,Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
47
|
Posma JJ, Grover SP, Hisada Y, Owens AP, Antoniak S, Spronk HM, Mackman N. Roles of Coagulation Proteases and PARs (Protease-Activated Receptors) in Mouse Models of Inflammatory Diseases. Arterioscler Thromb Vasc Biol 2019; 39:13-24. [PMID: 30580574 PMCID: PMC6310042 DOI: 10.1161/atvbaha.118.311655] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/19/2018] [Indexed: 12/20/2022]
Abstract
Activation of the blood coagulation cascade leads to fibrin deposition and platelet activation that are required for hemostasis. However, aberrant activation of coagulation can lead to thrombosis. Thrombi can cause tissue ischemia, and fibrin degradation products and activated platelets can enhance inflammation. In addition, coagulation proteases activate cells by cleavage of PARs (protease-activated receptors), including PAR1 and PAR2. Direct oral anticoagulants have recently been developed to specifically inhibit the coagulation proteases FXa (factor Xa) and thrombin. Administration of these inhibitors to wild-type mice can be used to determine the roles of FXa and thrombin in different inflammatory diseases. These results can be compared with the phenotypes of mice with deficiencies of either Par1 (F2r) or Par2 (F2rl1). However, inhibition of coagulation proteases will have effects beyond reducing PAR signaling, and a deficiency of PARs will abolish signaling from all proteases that activate these receptors. We will summarize studies that examine the roles of coagulation proteases, particularly FXa and thrombin, and PARs in different mouse models of inflammatory disease. Targeting FXa and thrombin or PARs may reduce inflammatory diseases in humans.
Collapse
Affiliation(s)
- Jens J Posma
- Laboratory for Clinical Thrombosis and Hemostasis, Departments of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Steven P Grover
- Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yohei Hisada
- Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - A. Phillip Owens
- Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, OH, USA
| | - Silvio Antoniak
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Henri M Spronk
- Laboratory for Clinical Thrombosis and Hemostasis, Departments of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Nigel Mackman
- Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
48
|
Bendstrup E, Wuyts W, Alfaro T, Chaudhuri N, Cornelissen R, Kreuter M, Melgaard Nielsen K, Münster AMB, Myllärniemi M, Ravaglia C, Vanuytsel T, Wijsenbeek M. Nintedanib in Idiopathic Pulmonary Fibrosis: Practical Management Recommendations for Potential Adverse Events. Respiration 2018; 97:173-184. [PMID: 30544129 DOI: 10.1159/000495046] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/31/2018] [Indexed: 11/19/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease with a dismal survival rate of only 3 years and no curative pharmacological therapy. The recent approval of 2 anti-fibrotic drugs (nintedanib and pirfenidone) that slow disease progression has provided some hope for patients. However, effectively managing anti-fibrotic treatment can be a challenge due to tolerability issues, the presence of pulmonary and extra-pulmonary comorbidities, and the need for concomitant medications in many patients. In general, making clear evidence-based decisions can be difficult for physicians because patients with comorbidities are often excluded from clinical trials. Since currently anti-fibrotic drugs are the only effective therapeutics capable of slowing disease progression, it is imperative that all treatment options are thoroughly evaluated and exhausted in each individual, irrespective of complicating factors, to permit the best outcome for the patient. In this review, we present data from clinical trials, post hoc analyses, post-marketing surveillance, and real-world studies that are relevant to the management of nintedanib treatment. In addition, we also provide practical recommendations developed by a multidisciplinary panel of experts for the management of nintedanib treatment in patients with IPF associated complications and those experiencing gastrointestinal side effects.
Collapse
Affiliation(s)
- Elisabeth Bendstrup
- Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark,
| | - Wim Wuyts
- Department of Respiratory Diseases, Interstitial Lung Disease and Lung Transplant Unit, University Hospitals Leuven, Leuven, Belgium.,KU Leuven, Department of Clinical and Experimental Medicine, Division of Respiratory Diseases, Laboratory of Respiratory Diseases, Lung Transplantation Unit, Leuven, Belgium
| | - Tiago Alfaro
- Pneumology Unit A, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Nazia Chaudhuri
- North West Interstitial Lung Disease Unit, Manchester University Foundation Trust, Wythenshawe, Manchester, United Kingdom
| | - Robin Cornelissen
- Department of Pulmonary Medicine, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Michael Kreuter
- Center for interstitial and rare lung diseases, Pneumology and respiratory critical care medicine, Thoraxklinik, University of Heidelberg, and Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | | | - Anna-Marie B Münster
- Unit of Thrombosis Research, Hospital of Southwest, University of Southern Denmark, Esbjerg, Denmark
| | - Marjukka Myllärniemi
- University of Helsinki and Helsinki University Hospital, Heart and Lung Center, Department of Pulmonary Medicine, Helsinki, Finland
| | - Claudia Ravaglia
- Department of Diseases of the Thorax, Ospedale GB Morgagni, Forlì, Italy
| | - Tim Vanuytsel
- Translational Research Center for Gastrointestinal Disorders (TARGID) and Department of Gastroenterology, University of Leuven, Leuven, Belgium
| | - Marlies Wijsenbeek
- Department of Pulmonary Disease, Erasmus Medical Centre, University Hospital Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
49
|
Bacha NC, Blandinieres A, Rossi E, Gendron N, Nevo N, Lecourt S, Guerin CL, Renard JM, Gaussem P, Angles-Cano E, Boulanger CM, Israel-Biet D, Smadja DM. Endothelial Microparticles are Associated to Pathogenesis of Idiopathic Pulmonary Fibrosis. Stem Cell Rev Rep 2018; 14:223-235. [PMID: 29101610 DOI: 10.1007/s12015-017-9778-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by obliteration of alveolar architecture, resulting in declining lung function and ultimately death. Pathogenic mechanisms remain unclear but involve a concomitant accumulation of scar tissue together with myofibroblasts activation. Microparticles (MPs) have been investigated in several human lung diseases as possible pathogenic elements, prognosis markers and therapeutic targets. We postulated that levels and cellular origins of circulating MPs might serve as biomarkers in IPF patients and/or as active players of fibrogenesis. Flow cytometry analysis showed a higher level of Annexin-V positive endothelial and platelet MPs in 41 IPF patients compared to 22 healthy volunteers. Moreover, in IPF patients with a low diffusing capacity of the lung for carbon monoxide (DLCO<40%), endothelial MPs (EMPs) were found significantly higher compared to those with DLCO>40% (p = 0.02). We then used EMPs isolated from endothelial progenitor cells (ECFCs) extracted from IPF patients or controls to modulate normal human lung fibroblast (NHLF) properties. We showed that EMPs did not modify proliferation, collagen deposition and myofibroblast transdifferentiation. However, EMPs from IPF patients stimulated migration capacity of NHLF. We hypothesized that this effect could result from EMPs fibrinolytic properties and found indeed higher plasminogen activation potential in total circulating MPs and ECFCs derived MPs issued from IPF patients compared to those isolated from healthy controls MPs. Our study showed that IPF is associated with an increased level of EMPs in the most severe patients, highlighting an active process of endothelial activation in the latter. Endothelial microparticles might contribute to the lung fibroblast invasion mediated, at least in part, by a fibrinolytic activity.
Collapse
Affiliation(s)
- Nour C Bacha
- Inserm UMR-S1140, Paris, France.,Sorbonne Paris Cite, Université Paris Descartes, Paris, France
| | - Adeline Blandinieres
- Inserm UMR-S1140, Paris, France.,Sorbonne Paris Cite, Université Paris Descartes, Paris, France.,Hematology Department and UMR-S1140, AP-HP, European Hospital Georges Pompidou, 20 rue Leblanc, 75015, Paris, France
| | - Elisa Rossi
- Inserm UMR-S1140, Paris, France.,Sorbonne Paris Cite, Université Paris Descartes, Paris, France
| | - Nicolas Gendron
- Inserm UMR-S1140, Paris, France.,Sorbonne Paris Cite, Université Paris Descartes, Paris, France.,Hematology Department and UMR-S1140, AP-HP, European Hospital Georges Pompidou, 20 rue Leblanc, 75015, Paris, France
| | - Nathalie Nevo
- Inserm UMR-S1140, Paris, France.,Sorbonne Paris Cite, Université Paris Descartes, Paris, France
| | | | - Coralie L Guerin
- National Cytometry Platform, Department of Infection and Immunity, Luxembourg Institute of Health, Luxembourg, France
| | - Jean Marie Renard
- Sorbonne Paris Cite, Université Paris Descartes, Paris, France.,Inserm UMR-S970, PARCC, Paris, France
| | - Pascale Gaussem
- Inserm UMR-S1140, Paris, France.,Sorbonne Paris Cite, Université Paris Descartes, Paris, France.,Hematology Department and UMR-S1140, AP-HP, European Hospital Georges Pompidou, 20 rue Leblanc, 75015, Paris, France
| | - Eduardo Angles-Cano
- Inserm UMR-S1140, Paris, France.,Sorbonne Paris Cite, Université Paris Descartes, Paris, France
| | - Chantal M Boulanger
- Sorbonne Paris Cite, Université Paris Descartes, Paris, France.,Inserm UMR-S970, PARCC, Paris, France
| | - Dominique Israel-Biet
- Inserm UMR-S1140, Paris, France.,Sorbonne Paris Cite, Université Paris Descartes, Paris, France.,Pneumology Department, AP-HP, European Hospital Georges Pompidou, Paris, France
| | - David M Smadja
- Inserm UMR-S1140, Paris, France. .,Sorbonne Paris Cite, Université Paris Descartes, Paris, France. .,Hematology Department and UMR-S1140, AP-HP, European Hospital Georges Pompidou, 20 rue Leblanc, 75015, Paris, France.
| |
Collapse
|
50
|
Korfei M, Stelmaszek D, MacKenzie B, Skwarna S, Chillappagari S, Bach AC, Ruppert C, Saito S, Mahavadi P, Klepetko W, Fink L, Seeger W, Lasky JA, Pullamsetti SS, Krämer OH, Guenther A. Comparison of the antifibrotic effects of the pan-histone deacetylase-inhibitor panobinostat versus the IPF-drug pirfenidone in fibroblasts from patients with idiopathic pulmonary fibrosis. PLoS One 2018; 13:e0207915. [PMID: 30481203 PMCID: PMC6258535 DOI: 10.1371/journal.pone.0207915] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 11/08/2018] [Indexed: 12/20/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease with a poor prognosis. Pirfenidone is the first antifibrotic agent to be approved for IPF-treatment as it is able to slow down disease progression. However, there is no curative treatment other than lung transplantation. Because epigenetic alterations are associated with IPF, histone deacetylase (HDAC)-inhibitors have recently been proven to attenuate fibrotic remodeling in vitro and in vivo. This study compared the effects of pirfenidone with the pan-HDAC-inhibitor panobinostat/LBH589, a FDA-approved drug for the treatment of multiple myeloma, head-to-head on survival, fibrotic activity and proliferation of primary IPF-fibroblasts in vitro. Methods Primary fibroblasts from six IPF-patients were incubated for 24h with vehicle (0.25% DMSO), panobinostat (LBH589, 85 nM) or pirfenidone (2.7 mM), followed by assessment of proliferation and expression analyses for profibrotic and anti-apoptosis genes, as well as for ER stress and apoptosis-markers. In addition, the expression status of all HDAC enzymes was examined. Results Treatment of IPF-fibroblasts with panobinostat or pirfenidone resulted in a downregulated expression of various extracellular matrix (ECM)-associated genes, as compared to vehicle-treated cells. In agreement, both drugs decreased protein level of phosphorylated (p)-STAT3, a transcription factor mediating profibrotic responses, in treated IPF-fibroblasts. Further, an increase in histone acetylation was observed in response to both treatments, but was much more pronounced and excessive in panobinostat-treated IPF-fibroblasts. Panobinostat, but not pirfenidone, led to a significant suppression of proliferation in IPF-fibroblasts, as indicated by WST1- and BrdU assay and markedly diminished levels of cyclin-D1 and p-histone H3. Furthermore, panobinostat-treatment enhanced α-tubulin-acetylation, decreased the expression of survival-related genes Bcl-XL and BIRC5/survivin, and was associated with induction of ER stress and apoptosis in IPF-fibroblasts. In contrast, pirfenidone-treatment maintained Bcl-XL expression, and was neither associated with ER stress-induction nor any apoptotic signaling. Pirfenidone also led to increased expression of HDAC6 and sirtuin-2, and enhanced α-tubulin-deacetylation. But in line with its ability to increase histone acetylation, pirfenidone reduced the expression of HDAC enzymes HDAC1, -2 and -9. Conclusions We conclude that, beside other antifibrotic mechanisms, pirfenidone reduces profibrotic signaling also through STAT3 inactivation and weak epigenetic alterations in IPF-fibroblasts, and permits survival of (altered) fibroblasts. The pan-HDAC-inhibitor panobinostat reduces profibrotic phenotypes while inducing cell cycle arrest and apoptosis in IPF-fibroblasts, thus indicating more efficiency than pirfenidone in inactivating IPF-fibroblasts. We therefore believe that HDAC-inhibitors such as panobinostat can present a novel therapeutic strategy for IPF.
Collapse
Affiliation(s)
- Martina Korfei
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- * E-mail:
| | - Daniel Stelmaszek
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - BreAnne MacKenzie
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Sylwia Skwarna
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Shashipavan Chillappagari
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Anna C. Bach
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Clemens Ruppert
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Giessen, Germany
| | - Shigeki Saito
- Department of Medicine, Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Poornima Mahavadi
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Walter Klepetko
- Department of Thoracic Surgery, Vienna General Hospital, Vienna, Austria
- European IPF Network and European IPF Registry, Giessen, Germany
| | - Ludger Fink
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Giessen, Germany
- Institute of Pathology and Cytology, Wetzlar, Germany
| | - Werner Seeger
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Giessen, Germany
- Max-Planck-Institute for Heart and Lung Research, Department of Lung Development and Remodeling, Bad Nauheim, Germany
| | - Joseph A. Lasky
- Department of Medicine, Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Soni S. Pullamsetti
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Max-Planck-Institute for Heart and Lung Research, Department of Lung Development and Remodeling, Bad Nauheim, Germany
| | - Oliver H. Krämer
- Department of Toxicology, University Medical Center, Mainz, Germany
| | - Andreas Guenther
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Giessen, Germany
- European IPF Network and European IPF Registry, Giessen, Germany
- Agaplesion Lung Clinic Waldhof Elgershausen, Greifenstein, Germany
| |
Collapse
|