1
|
Zhang X, Yuan Z, Shi X, Yang J. Targeted therapy for idiopathic pulmonary fibrosis: a bibliometric analysis of 2004-2024. Front Med (Lausanne) 2025; 12:1543571. [PMID: 40182841 PMCID: PMC11967194 DOI: 10.3389/fmed.2025.1543571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a progressive and irreversible interstitial lung disease characterized by high mortality rates. An expanding body of evidence highlights the critical role of targeted therapies in the management of IPF. Nevertheless, there is a paucity of bibliometric studies that have comprehensively assessed this domain. This study seeks to examine global literature production and research trends related to targeted therapies for IPF. Method A literature search was conducted using the Web of Science Core Collection, encompassing publications from 2004 to 2024, focusing on targeted therapies for IPF. The bibliometric analysis utilized tools such as VOSviewer, CiteSpace, and the "bibliometrix" package in R. Results A total of 2,779 papers were included in the analysis, demonstrating a general trend of continuous growth in the number of publications over time. The United States contributed the highest number of publications, totaling 1,052, while France achieved the highest average citation rate at 75.74. The University of Michigan Medical School was the leading institution in terms of publication output, with 88 papers. Principal Investigator Naftali Kaminski was identified as the most prolific researcher in the field. The American Journal of Respiratory Cell and Molecular Biology emerged as the journal with the highest number of publications, featuring 98 articles. In recent years, the research has emerged surrounding targeted therapies for IPF, particularly focusing on agents such as TGF-β, pathogenesis, and autotaxin inhibitor. Conclusion In this bibliometric study, we systematically analyze research trends related to targeted therapies for IPF, elucidating recent research frontiers and emerging directions. The selected keywords-idiopathic pulmonary fibrosis, targeted therapy, bibliometric analysis, transforming growth factor β, and autotaxin inhibitor-capture the essential aspects of this research domain. This analysis serves as a reference point for future investigations into targeted therapies.
Collapse
Affiliation(s)
- Xinlei Zhang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou, Zhejiang, China
| | - Zengze Yuan
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiawei Shi
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou, Zhejiang, China
| | - Junchao Yang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Farooq M, Hwang M, Khan AW, Batool M, Ahmad B, Kim W, Kim MS, Choi S. Identification of a novel fibroblast growth factor receptor-agonistic peptide and its effect on diabetic wound healing. Life Sci 2025; 364:123432. [PMID: 39884341 DOI: 10.1016/j.lfs.2025.123432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
AIMS Fibroblast growth factor (FGF) is a broad class of secretory chemicals that act via FGF receptors (FGFR). The study aims to explore the role of a novel peptide, FAP1 (FGFR-agonistic peptide 1), in tissue regeneration and repair. It investigates whether FAP1 mimics basic fibroblast growth factor (bFGF) and accelerates wound healing both in vitro and in vivo. MAIN METHODS In this study, a novel peptide was designed and its ability to mimic bFGF was assessed through different in vitro experiments including its effect on cell proliferation, wound healing, cell signaling including FGFR1 phosphorylation and activation of mitogen-activated protein kinases (MAPKs). Specificity was confirmed through surface plasmon resonance (SPR) analysis and co-treatment with FGFR inhibitor, erdafitinib. In vivo, the effect of FAP1 on diabetic wound healing was tested in a mouse model, examining collagen production and the migration and proliferation of keratinocytes and fibroblasts. KEY FINDINGS FAP1 specifically phosphorylated FGFR and activated MAPKs similar to bFGF. In vitro, it induced cell proliferation and accelerated wound healing. In vivo, FAP1 improved diabetic wound healing by increasing collagen production and promoting keratinocyte and fibroblast migration and proliferation. The specificity of FAP1 was confirmed through SPR. SIGNIFICANCE FAP1 shows potential as a novel pharmacological alternative to natural bFGF for skin tissue regeneration and repair. Its ability to accelerate wound healing and its specificity for FGFR suggest that FAP1 could serve as a cost-effective substitute for bFGF protein in therapeutic applications.
Collapse
Affiliation(s)
- Mariya Farooq
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongton-gu, Suwon 16502, Republic of Korea; Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Moonjung Hwang
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Abdul Waheed Khan
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Maria Batool
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongton-gu, Suwon 16502, Republic of Korea
| | - Bilal Ahmad
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongton-gu, Suwon 16502, Republic of Korea
| | - Wook Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea.
| | - Sangdun Choi
- S&K Therapeutics, Ajou University Campus Plaza 418, Worldcup-ro 199, Yeongton-gu, Suwon 16502, Republic of Korea.
| |
Collapse
|
3
|
Nickle A, Ko S, Merrill AE. Fibroblast growth factor 2. Differentiation 2024; 139:100733. [PMID: 37858405 PMCID: PMC11009566 DOI: 10.1016/j.diff.2023.10.001] [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: 05/22/2023] [Revised: 09/20/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023]
Abstract
Fibroblast Growth Factor 2 (FGF2), also known as basic fibroblast growth factor, is a potent stimulator of growth and differentiation in multiple tissues. Its discovery traces back over 50 years ago when it was first isolated from bovine pituitary extracts due to its ability to stimulate fibroblast proliferation. Subsequent studies investigating the genomic structure of FGF2 identified multiple protein isoforms, categorized as the low molecular weight and high molecular weight FGF2. These isoforms arise from alternative translation initiation events and exhibit unique molecular and cellular functions. In this concise review, we aim to provide an overview of what is currently known about the structure, expression, and functions of the FGF2 isoforms within the contexts of development, homeostasis, and disease.
Collapse
Affiliation(s)
- Audrey Nickle
- Center for Craniofacial Molecular Biology, Department of Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Sebastian Ko
- Center for Craniofacial Molecular Biology, Department of Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Amy E Merrill
- Center for Craniofacial Molecular Biology, Department of Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
4
|
Olave NC, Halloran B, Ambalavanan N. FGF2 is secreted in extracellular vesicles from lung cells. Am J Physiol Lung Cell Mol Physiol 2024; 327:L359-L370. [PMID: 39010825 PMCID: PMC11444508 DOI: 10.1152/ajplung.00225.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 06/11/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
The 18-kDa isoform of basic fibroblast growth factor (bFGF/FGF2) lacks a conventional signal peptide sequence and is exported by a novel membrane-associated transport pathway. Extracellular vesicles (EVs) are increasingly recognized as mediators of intercellular communication in the lung, and our prior work demonstrates that EVs carry cargo that contributes to hyperoxic lung injury and are biomarkers for bronchopulmonary dysplasia. We used primary human bronchial epithelial (HBE), pulmonary artery endothelial (HPAE), and fibroblast (HNF) cells to determine whether FGF2 was secreted in EVs. EVs were isolated by ultracentrifugation from HBE, HPAE, and HNF exposed to either normoxia or hyperoxia, followed by nanoparticle tracking analysis and electron microscopy. Hyperoxia exposure increased the total EV number. All three cell types released FGF2-18kDa both directly into the extracellular environment (secretome), as well as in EVs. HBE released more FGF2-18kDa in EVs during hyperoxia, and these were internalized and localized to both nuclei and cytoplasm of recipient cells. By co-immunoprecipitation, we identified potential binding partners of FGF2-18kDa in the nuclei, including histone 1.2 (H1.2) binding protein, that may mediate downstream effects that do not involve FGF2 binding to cell surface receptors. FGF2-18kDa interaction with H1.2 binding protein may indicate a mechanism by which FGF2 secreted in EVs modulates cellular processes. FGF2 was also found to increase angiogenesis by Matrigel assay. Further studies are necessary to determine the biological relevance of FGF2 in EVs as modulators of lung injury and disease.NEW & NOTEWORTHY We found that multiple lung cell types release basic fibroblast growth factor (FGF2)-18kDa both directly into the extracellular environment (secretome), as well as in extracellular vesicles (EVs). Bronchial epithelial cells released more FGF2-18kDa in EVs during hyperoxia, which could be internalized rapidly by recipient cells. We also identified potential binding partners of FGF2-18kDa in nuclei that may mediate downstream effects that do not involve FGF2 binding to cell surface receptors. We also confirmed a potential angiogenic role for FGF2-18kDa.
Collapse
Affiliation(s)
- Nelida C Olave
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Brian Halloran
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Namasivayam Ambalavanan
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
5
|
Chang D, Dela Cruz C, Sharma L. Beneficial and Detrimental Effects of Cytokines during Influenza and COVID-19. Viruses 2024; 16:308. [PMID: 38400083 PMCID: PMC10892676 DOI: 10.3390/v16020308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Cytokines are signaling molecules that play a role in myriad processes, including those occurring during diseases and homeostasis. Their homeostatic function begins during embryogenesis and persists throughout life, including appropriate signaling for the cell and organism death. During viral infections, antiviral cytokines such as interferons and inflammatory cytokines are upregulated. Despite the well-known benefits of these cytokines, their levels often correlate with disease severity, linking them to unfavorable outcomes. In this review, we discuss both the beneficial and pathological functions of cytokines and the potential challenges in separating these two roles. Further, we discuss challenges in targeting these cytokines during disease and propose a new method for quantifying the cytokine effect to limit the pathological consequences while preserving their beneficial effects.
Collapse
Affiliation(s)
- De Chang
- College of Pulmonary and Critical Care Medicine of Eighth Medical Center, Chinese PLA General Hospital, Beijing 100028, China;
- Department of Pulmonary and Critical Care Medicine of Seventh Medical Center, Chinese PLA General Hospital, Beijing 100028, China
| | - Charles Dela Cruz
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Lokesh Sharma
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| |
Collapse
|
6
|
Cates WT, Denbeigh JM, Salvagno RT, Kakar S, van Wijnen AJ, Eaton C. Inflammatory Markers Involved in the Pathogenesis of Dupuytren's Contracture. Crit Rev Eukaryot Gene Expr 2024; 34:1-35. [PMID: 38912961 DOI: 10.1615/critreveukaryotgeneexpr.2024052889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Dupuytren's disease is a common fibroproliferative disease that can result in debilitating hand deformities. Partial correction and return of deformity are common with surgical or clinical treatments at present. While current treatments are limited to local procedures for relatively late effects of the disease, the pathophysiology of this connective tissue disorder is associated with both local and systemic processes (e.g., fibrosis, inflammation). Hence, a better understanding of the systemic circulation of Dupuytren related cytokines and growth factors may provide important insights into disease progression. In addition, systemic biomarker analysis could yield new concepts for treatments of Dupuytren that attenuate circulatory factors (e.g., anti-inflammatory agents, neutralizing antibodies). Progress in the development of any disease modifying biologic treatment for Dupuytren has been hampered by the lack of clinically useful biomarkers. The characterization of nonsurgical Dupuytren biomarkers will permit disease staging from diagnostic and prognostic perspectives, as well as allows evaluation of biologic responses to treatment. Identification of such markers may transcend their use in Dupuytren treatment, because fibrotic biological processes fundamental to Dupuytren are relevant to fibrosis in many other connective tissues and organs with collagen-based tissue compartments. There is a wide range of potential Dupuytren biomarker categories that could be informative, including disease determinants linked to genetics, collagen metabolism, as well as immunity and inflammation (e.g., cytokines, chemokines). This narrative review provides a broad overview of previous studies and emphasizes the importance of inflammatory mediators as candidate circulating biomarkers for monitoring Dupuytren's disease.
Collapse
Affiliation(s)
- William T Cates
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Janet M Denbeigh
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Sanjeev Kakar
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Andre J van Wijnen
- Department of Biochemistry, University of Vermont, Burlington, VT 05405, USA
| | | |
Collapse
|
7
|
Takahashi N, Nakashima R, Nasu A, Hayashi M, Fujikawa H, Kawakami T, Eto Y, Kishimoto T, Fukuyama A, Ogasawara C, Kawano K, Fujiwara Y, Suico MA, Kai H, Shuto T. T 3 Intratracheal Therapy Alleviates Pulmonary Pathology in an Elastase-Induced Emphysema-Dominant COPD Mouse Model. Antioxidants (Basel) 2023; 13:30. [PMID: 38247455 PMCID: PMC10812479 DOI: 10.3390/antiox13010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 01/23/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a complex pulmonary condition characterized by bronchitis, emphysema, and mucus stasis. Due to the variability in symptoms among patients, traditional approaches to treating COPD as a singular disease are limited. This led us to focus on phenotype/endotype classifications. In this study, we explore the potential therapeutic role of thyroid hormone (T3) by using mouse models: emphysema-dominant elastase-induced COPD and airway-dominant C57BL/6-βENaC-Tg to represent different types of the disease. Here, we showed that intratracheal T3 treatment (40, 80 μg/kg, i.t., every other day) resulted in significant improvements regarding emphysema and the enhancement of respiratory function in the elastase-induced COPD model. T3-dependent improvement is likely linked to the up-regulation of Ppargc1a, a master regulator of mitochondrial biogenesis, and Gclm, a factor associated with oxidative stress. Conversely, neither short- nor long-term T3 treatments improved COPD pathology in the C57BL/6-βENaC-Tg mice. Because the up-regulation of extrathyroidal T3-producing enzyme Dio2, which is also considered a marker of T3 requirement, was specifically observed in elastase-induced COPD lungs, these results demonstrate that exogenous T3 supplementation may have therapeutic potential for acute but not chronic COPD exacerbation. Moreover, this study highlights the relevance of considering not only COPD phenotypes but also COPD endotypes (expression levels of Ppargc1a and/or Dio2) in the research and development of better treatment approaches for COPD.
Collapse
Affiliation(s)
- Noriki Takahashi
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (N.T.); (A.N.); (M.H.); (H.F.); (T.K.); (T.K.); (A.F.); (C.O.); (K.K.); (M.A.S.); (H.K.)
- Program for Leading Graduate Schools “HIGO (Health Life Science: Interdisciplinary and Global Oriented) Program”, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Ryunosuke Nakashima
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (N.T.); (A.N.); (M.H.); (H.F.); (T.K.); (T.K.); (A.F.); (C.O.); (K.K.); (M.A.S.); (H.K.)
| | - Aoi Nasu
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (N.T.); (A.N.); (M.H.); (H.F.); (T.K.); (T.K.); (A.F.); (C.O.); (K.K.); (M.A.S.); (H.K.)
- Program for Leading Graduate Schools “HIGO (Health Life Science: Interdisciplinary and Global Oriented) Program”, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Megumi Hayashi
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (N.T.); (A.N.); (M.H.); (H.F.); (T.K.); (T.K.); (A.F.); (C.O.); (K.K.); (M.A.S.); (H.K.)
| | - Haruka Fujikawa
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (N.T.); (A.N.); (M.H.); (H.F.); (T.K.); (T.K.); (A.F.); (C.O.); (K.K.); (M.A.S.); (H.K.)
- Program for Leading Graduate Schools “HIGO (Health Life Science: Interdisciplinary and Global Oriented) Program”, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Taisei Kawakami
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (N.T.); (A.N.); (M.H.); (H.F.); (T.K.); (T.K.); (A.F.); (C.O.); (K.K.); (M.A.S.); (H.K.)
| | - Yuka Eto
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (N.T.); (A.N.); (M.H.); (H.F.); (T.K.); (T.K.); (A.F.); (C.O.); (K.K.); (M.A.S.); (H.K.)
| | - Tomoki Kishimoto
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (N.T.); (A.N.); (M.H.); (H.F.); (T.K.); (T.K.); (A.F.); (C.O.); (K.K.); (M.A.S.); (H.K.)
| | - Ayami Fukuyama
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (N.T.); (A.N.); (M.H.); (H.F.); (T.K.); (T.K.); (A.F.); (C.O.); (K.K.); (M.A.S.); (H.K.)
| | - Choyo Ogasawara
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (N.T.); (A.N.); (M.H.); (H.F.); (T.K.); (T.K.); (A.F.); (C.O.); (K.K.); (M.A.S.); (H.K.)
| | - Keisuke Kawano
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (N.T.); (A.N.); (M.H.); (H.F.); (T.K.); (T.K.); (A.F.); (C.O.); (K.K.); (M.A.S.); (H.K.)
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1, Honjo, Kumamoto Chuo-ku, Kumamoto 860-8556, Japan;
| | - Mary Ann Suico
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (N.T.); (A.N.); (M.H.); (H.F.); (T.K.); (T.K.); (A.F.); (C.O.); (K.K.); (M.A.S.); (H.K.)
- Global Center for Natural Resources Sciences, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Hirofumi Kai
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (N.T.); (A.N.); (M.H.); (H.F.); (T.K.); (T.K.); (A.F.); (C.O.); (K.K.); (M.A.S.); (H.K.)
- Global Center for Natural Resources Sciences, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Tsuyoshi Shuto
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; (N.T.); (A.N.); (M.H.); (H.F.); (T.K.); (T.K.); (A.F.); (C.O.); (K.K.); (M.A.S.); (H.K.)
- Global Center for Natural Resources Sciences, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-Honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| |
Collapse
|
8
|
Jang S, Lee H, Park J, Cha SR, Lee J, Park Y, Jang SH, Park JR, Hong SH, Yang SR. PTD-FGF2 Attenuates Elastase Induced Emphysema in Mice and Alveolar Epithelial Cell Injury. COPD 2023; 20:109-118. [PMID: 36882376 DOI: 10.1080/15412555.2023.2174842] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Aberrant communication in alveolar epithelium is a major feature of inflammatory response for the airway remodeling leading to chronic obstructive pulmonary disease (COPD). In this study, we investigated the effect of protein transduction domains (PTD) conjugated Basic Fibroblast Growth Factor (FGF2) (PTD-FGF2) in response to cigarette smoke extract (CSE) in MLE-12 cells and porcine pancreatic elastase (PPE)-induced emphysematous mice. When PPE-induced mice were intraperitoneally treated with 0.1-0.5 mg/kg PTD-FGF2 or FGF2, the linear intercept, infiltration of inflammatory cells into alveoli and pro-inflammatory cytokines were significantly decreased. In western blot analysis, phosphorylated protein levels of c-Jun N-terminal Kinase 1/2 (JNK1/2), extracellular signal-regulated kinase (ERK1/2) and p38 mitogen-activated protein kinases (MAPK) were decreased in PPE-induced mice treated PTD-FGF2. In MLE-12 cells, PTD-FGF2 treatment decreased reactive oxygen species (ROS) production and further decreased Interleukin-6 (IL-6) and IL-1b cytokines in response to CSE. In addition, phosphorylated protein levels of ERK1/2, JNK1/2 and p38 MAPK were reduced. We next determined microRNA expression in the isolated exosomes of MLE-12 cells. In reverse transcription-polymerase chain reaction (RT-PCR) analysis, level of let-7c miRNA was significantly increased while levels of miR-9 and miR-155 were decreased in response to CSE. These data suggest that PTD-FGF2 treatment plays a protective role in regulation of let-7c, miR-9 and miR-155 miRNA expressions and MAPK signaling pathways in CSE-induced MLE-12 cells and PPE-induced emphysematous mice.
Collapse
Affiliation(s)
- Soojin Jang
- Department of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Hanbyeol Lee
- Department of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jaehyun Park
- Department of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Sang-Ryul Cha
- Department of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jooyeon Lee
- Department of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Youngheon Park
- Department of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Sang Ho Jang
- Bioceltran Co., Ltd, Chuncheon, Republic of Korea
| | - Jeong-Ran Park
- Department of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Se-Ran Yang
- Department of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| |
Collapse
|
9
|
Tian X, Jia Y, Guo Y, Liu H, Cai X, Li Y, Tian Z, Sun C. Fibroblast growth factor 2 acts as an upstream regulator of inhibition of pulmonary fibroblast activation. FEBS Open Bio 2023; 13:1895-1909. [PMID: 37583315 PMCID: PMC10549223 DOI: 10.1002/2211-5463.13691] [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: 03/06/2023] [Revised: 07/06/2023] [Accepted: 08/11/2023] [Indexed: 08/17/2023] Open
Abstract
Fibroblast growth factor (FGF) signaling plays a crucial role in lung development and repair. Fibroblast growth factor 2 (FGF2) can inhibit fibrotic gene expression and suppress the differentiation of pulmonary fibroblasts (PFs) into myofibroblasts in vitro, suggesting that FGF2 is a potential target for inhibiting pulmonary fibrosis. To gain deeper insights into the molecular mechanism underlying FGF2-mediated regulation of PFs, we performed mRNA sequencing analysis to systematically and globally uncover the regulated genes and biological functions of FGF2 in PFs. Gene Ontology analysis revealed that the differentially expressed genes regulated by FGF2 were enriched in multiple cellular functions including extracellular matrix (ECM) organization, cytoskeleton formation, β-catenin-independent Wnt signaling pathway, supramolecular fiber organization, epithelial cell proliferation, and cell adhesion. Gene Set Enrichment Analysis and cellular experiments confirmed that FGF2 can suppress ECM and actin filament organization and increase PFs proliferation. Taken together, these findings indicate that FGF2 acts as an upstream regulator of the inhibition of PFs activation and may play a regulatory role in pulmonary fibrosis.
Collapse
Affiliation(s)
- Xiangqin Tian
- Henan Key Laboratory of Medical Tissue RegenerationXinxiang Medical UniversityChina
| | - Yangyang Jia
- Henan Key Laboratory of Medical Tissue RegenerationXinxiang Medical UniversityChina
| | - Yonglong Guo
- Department of Cardiology, The First Affiliated HospitalXinxiang Medical UniversityChina
| | - Hongyin Liu
- Henan Key Laboratory of Medical Tissue RegenerationXinxiang Medical UniversityChina
| | - Xinhua Cai
- Henan Key Laboratory of Medical Tissue RegenerationXinxiang Medical UniversityChina
| | - Yong Li
- Department of Biochemistry, Institute of Systems, Molecular and Integrative BiologyUniversity of LiverpoolUK
| | - Zhuangzhuang Tian
- Henan Key Laboratory of Medical Tissue RegenerationXinxiang Medical UniversityChina
| | - Changye Sun
- Henan Key Laboratory of Medical Tissue RegenerationXinxiang Medical UniversityChina
| |
Collapse
|
10
|
Spector C, De Sanctis CM, Panettieri RA, Koziol-White CJ. Rhinovirus induces airway remodeling: what are the physiological consequences? Respir Res 2023; 24:238. [PMID: 37773065 PMCID: PMC10540383 DOI: 10.1186/s12931-023-02529-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Rhinovirus infections commonly evoke asthma exacerbations in children and adults. Recurrent asthma exacerbations are associated with injury-repair responses in the airways that collectively contribute to airway remodeling. The physiological consequences of airway remodeling can manifest as irreversible airway obstruction and diminished responsiveness to bronchodilators. Structural cells of the airway, including epithelial cells, smooth muscle, fibroblasts, myofibroblasts, and adjacent lung vascular endothelial cells represent an understudied and emerging source of cellular and extracellular soluble mediators and matrix components that contribute to airway remodeling in a rhinovirus-evoked inflammatory environment. MAIN BODY While mechanistic pathways associated with rhinovirus-induced airway remodeling are still not fully characterized, infected airway epithelial cells robustly produce type 2 cytokines and chemokines, as well as pro-angiogenic and fibroblast activating factors that act in a paracrine manner on neighboring airway cells to stimulate remodeling responses. Morphological transformation of structural cells in response to rhinovirus promotes remodeling phenotypes including induction of mucus hypersecretion, epithelial-to-mesenchymal transition, and fibroblast-to-myofibroblast transdifferentiation. Rhinovirus exposure elicits airway hyperresponsiveness contributing to irreversible airway obstruction. This obstruction can occur as a consequence of sub-epithelial thickening mediated by smooth muscle migration and myofibroblast activity, or through independent mechanisms mediated by modulation of the β2 agonist receptor activation and its responsiveness to bronchodilators. Differential cellular responses emerge in response to rhinovirus infection that predispose asthmatic individuals to persistent signatures of airway remodeling, including exaggerated type 2 inflammation, enhanced extracellular matrix deposition, and robust production of pro-angiogenic mediators. CONCLUSIONS Few therapies address symptoms of rhinovirus-induced airway remodeling, though understanding the contribution of structural cells to these processes may elucidate future translational targets to alleviate symptoms of rhinovirus-induced exacerbations.
Collapse
Affiliation(s)
- Cassandra Spector
- Rutgers Institute for Translation Medicine and Science, New Brunswick, NJ, USA
| | - Camden M De Sanctis
- Rutgers Institute for Translation Medicine and Science, New Brunswick, NJ, USA
| | | | | |
Collapse
|
11
|
Adegunsoye A, Baccile R, Best TJ, Zaksas V, Zhang H, Karnik R, Patel BK, Solomonides AE, Parker WF, Solway J. Pharmacotherapy and pulmonary fibrosis risk after SARS-CoV-2 infection: a prospective nationwide cohort study in the United States. LANCET REGIONAL HEALTH. AMERICAS 2023; 25:100566. [PMID: 37564420 PMCID: PMC10410516 DOI: 10.1016/j.lana.2023.100566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/06/2023] [Accepted: 07/19/2023] [Indexed: 08/12/2023]
Abstract
Background Pulmonary fibrosis is characterized by lung parenchymal destruction and can increase morbidity and mortality. Pulmonary fibrosis commonly occurs following hospitalization for SARS-CoV-2 infection. As there are medications that modify pulmonary fibrosis risk, we investigated whether distinct pharmacotherapies (amiodarone, cancer chemotherapy, corticosteroids, and rituximab) are associated with differences in post-COVID-19 pulmonary fibrosis incidence. Methods We used the National COVID-19 Cohort Collaboration (N3C) Data Enclave, which aggregates and harmonizes COVID-19 data across the United States, to assess pulmonary fibrosis incidence documented at least 60 days after COVID-19 diagnosis among adults hospitalized between January 1st, 2020 and July 6th, 2022 without pre-existing pulmonary fibrosis. We used propensity scores to match pre-COVID-19 drug-exposed and unexposed cohorts (1:1) based on covariates with known influence on pulmonary fibrosis incidence, and estimated the association of drug exposure with risk for post-COVID-19 pulmonary fibrosis. Sensitivity analyses considered pulmonary fibrosis incidence documented at least 30- or 90-days post-hospitalization and pulmonary fibrosis incidence in the COVID-19-negative N3C population. Findings Among 5,923,394 patients with COVID-19, we analyzed 452,951 hospitalized adults, among whom pulmonary fibrosis incidence was 1.1 per 100-person-years. 277,984 hospitalized adults with COVID-19 were included in our primary analysis, among whom all drug exposed cohorts were well-matched to unexposed cohorts (standardized mean differences <0.1). The post-COVID-19 pulmonary fibrosis incidence rate ratio (IRR) was 2.5 (95% CI 1.2-5.1, P = 0.01) for rituximab, 1.6 (95% CI 1.3-2.0, P < 0.0001) for chemotherapy, and 1.2 (95% CI 1.0-1.3, P = 0.02) for corticosteroids. Amiodarone exposure had no significant association with post-COVID-19 pulmonary fibrosis (IRR = 0.8, 95% CI 0.6-1.1, P = 0.24). In sensitivity analyses, pre-COVID-19 corticosteroid use was not consistently associated with post-COVID-19 pulmonary fibrosis. In the COVID-19 negative hospitalized population (n = 1,240,461), pulmonary fibrosis incidence was lower overall (0.6 per 100-person-years) and for patients exposed to all four drugs. Interpretation Recent rituximab or cancer chemotherapy before COVID-19 infection in hospitalized patients is associated with increased risk for post-COVID-19 pulmonary fibrosis. Funding The analyses described in this publication were conducted with data or tools accessed through the NCATS N3C Data Enclave https://covid.cd2h.org and N3C Attribution & Publication Policy v1.2-2020-08-25b supported by NIHK23HL146942, NIHK08HL150291, NIHK23HL148387, NIHUL1TR002389, NCATSU24 TR002306, and a SECURED grant from the Walder Foundation/Center for Healthcare Delivery Science and Innovation, University of Chicago. WFP received a grant from the Greenwall Foundation. This research was possible because of the patients whose information is included within the data and the organizations (https://ncats.nih.gov/n3c/resources/data-contribution/data-transfer-agreement-signatories) and scientists who have contributed to the on-going development of this community resource (https://doi.org/10.1093/jamia/ocaa196).
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA
- Committee on Clinical Pharmacology & Pharmacogenomics, The University of Chicago, Chicago, IL, USA
| | - Rachel Baccile
- Center for Health and the Social Sciences, The University of Chicago, Chicago, IL, USA
| | - Thomas J. Best
- Center for Health and the Social Sciences, The University of Chicago, Chicago, IL, USA
| | - Victoria Zaksas
- Center for Translational Data Science, The University of Chicago, Chicago, IL, USA
- Clever Research Lab, Springfield, IL, USA
| | - Hui Zhang
- Center for Health and the Social Sciences, The University of Chicago, Chicago, IL, USA
| | - Rasika Karnik
- Section of General Internal Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Bhakti K. Patel
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Anthony E. Solomonides
- Outcomes Research Network, Research Institute, NorthShore University HealthSystem, Evanston, IL, USA
- The Institute for Translational Medicine, University of Chicago, Chicago, IL, USA
| | - William F. Parker
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Julian Solway
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA
- The Institute for Translational Medicine, University of Chicago, Chicago, IL, USA
| | - N3C Consortium
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA
- Committee on Clinical Pharmacology & Pharmacogenomics, The University of Chicago, Chicago, IL, USA
- Center for Health and the Social Sciences, The University of Chicago, Chicago, IL, USA
- Center for Translational Data Science, The University of Chicago, Chicago, IL, USA
- Clever Research Lab, Springfield, IL, USA
- Section of General Internal Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA
- Outcomes Research Network, Research Institute, NorthShore University HealthSystem, Evanston, IL, USA
- The Institute for Translational Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
12
|
Wang X, Hou X, Zhao Y, Zhao R, Dai J, Dai H, Wang C. The early and late intervention effects of collagen-binding FGF2 on elastase-induced lung injury. Biomed Pharmacother 2023; 158:114147. [PMID: 36584430 DOI: 10.1016/j.biopha.2022.114147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/05/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Chronic obstructive pulmonary disease (COPD) has high morbidity and mortality, with no effective treatment at present. Emphysema, a major component of COPD, is a leading cause of human death worldwide. Fibroblast growth factor 2 (FGF2) is implicated in the pathogenesis of pulmonary emphysema and may play an important role in the lung repair process after injury, but concerns remain with respect to its effectiveness. OBJECTIVE In the present work, we sought to determine how the timing (early and late intervention) of sustained-release FGF2 system administration impacted its effectiveness on a porcine pancreatic elastase (PPE)-induced lung injury mouse model. METHODS To examine the early intervention efficiency of collagen-binding FGF2 (CBD-FGF2), mice received intratracheally nebulized CBD-FGF2 with concurrent intratracheal injection of PPE. To explore the late intervention effect, CBD-FGF2 was intratracheally aerosolized after PPE administration, and lungs were collected after CBD-FGF2 treatment for subsequent analysis. RESULT In response to PPE, mice had significantly increased alveolar diameter, collagen deposition and expression of inflammatory factors and decreased lung function indices and expression of alveolar epithelium markers. Our results indicate that CBD-FGF2 administration was able to prevent and repair elastase-induced lung injury partly through the suppression of the inflammatory response and recovery of the alveolar epithelium. The early use of CBD-FGF2 for the prevention of PPE-induced emphysema showed better results than late therapeutic administration against established emphysema. CONCLUSION These data provide insight regarding the prospective role of a drug-based option (CBD-FGF2) for preventing and curing emphysema.
Collapse
Affiliation(s)
- Xin Wang
- Beijing University of Chinese Medicine, Beijing 100029, China; Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Peking Union Medical College, Beijing 100029, China
| | - Xianglin Hou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ruiming Zhao
- Beijing University of Chinese Medicine, Beijing 100029, China; Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Peking Union Medical College, Beijing 100029, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Peking Union Medical College, Beijing 100029, China; National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing 100029, China.
| | - Chen Wang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Peking Union Medical College, Beijing 100029, China; National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing 100029, China.
| |
Collapse
|
13
|
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
|
14
|
Luo Y, Deng D, Lin L, Zhou Y, Wang L, Zou X, Wang X. FGF2 isoforms play distinct roles in tubular epithelial-to-mesenchymal transition in diabetic nephropathy. Exp Cell Res 2022; 420:113355. [PMID: 36115414 DOI: 10.1016/j.yexcr.2022.113355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 08/17/2022] [Accepted: 09/09/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The role of different isoforms of Fibroblast growth factor-2 (FGF2) in tubular epithelial-to-mesenchymal transition (EMT) in diabetic nephropathy remains unknown. We aimed to evaluate the role of FGF2 isoforms in the pathogenesis of EMT. MATERIALS AND METHODS Western blot and immunofluorescence were used to assess the expression of FGF2 isoforms in db/db mice and high glucose-stimulated HK2 cells. The effects of specific FGF2 isoforms on EMT were explored via overexpression or knockdown of the corresponding isoform in HK2 cells cultivated in high glucose. RESULTS Expression of low molecular weight (LMW) FGF2 was up-regulated while high molecular weight (HMW) FGF2 was down-regulated in the kidney of db/db mice and HK2 cells cultured in high glucose that underwent EMT. Overexpression of the LMW FGF2 enhanced EMT changes, while overexpression of the HMW FGF2 attenuated EMT. Knockdown of HMW FGF2 in HK2 cells promoted the EMT process. CONCLUSIONS The expression and function of LMW and HMW FGF2 differed in the process of EMT in tubular cells. LMW FGF2 contributed to EMT, while HMW FGF2 played a protective role in the EMT process.
Collapse
Affiliation(s)
- Yingying Luo
- School of Clinical Medicine, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Danfang Deng
- Department of Nephrology, Hubei Provincial Hospital of Chinese Medicine, The Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China; Department of Nephrology, Hubei Provincial Traditional Chinese Medicine Research Institute, Wuhan, 430074, China; Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Wuhan, 430074, China
| | - Lamei Lin
- Department of Nephrology, Hubei Provincial Hospital of Chinese Medicine, The Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China; Department of Nephrology, Hubei Provincial Traditional Chinese Medicine Research Institute, Wuhan, 430074, China; Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Wuhan, 430074, China
| | - Yikun Zhou
- School of Clinical Medicine, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Lan Wang
- Department of Nephrology, Hubei Provincial Hospital of Chinese Medicine, The Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China; Department of Nephrology, Hubei Provincial Traditional Chinese Medicine Research Institute, Wuhan, 430074, China; Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Wuhan, 430074, China
| | - Xinrong Zou
- Department of Nephrology, Hubei Provincial Hospital of Chinese Medicine, The Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China; Department of Nephrology, Hubei Provincial Traditional Chinese Medicine Research Institute, Wuhan, 430074, China; Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Wuhan, 430074, China
| | - Xiaoqin Wang
- Department of Nephrology, Hubei Provincial Hospital of Chinese Medicine, The Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China; Department of Nephrology, Hubei Provincial Traditional Chinese Medicine Research Institute, Wuhan, 430074, China; Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Wuhan, 430074, China.
| |
Collapse
|
15
|
Sun C, Tian X, Jia Y, Yang M, Li Y, Fernig DG. Functions of exogenous FGF signals in regulation of fibroblast to myofibroblast differentiation and extracellular matrix protein expression. Open Biol 2022; 12:210356. [PMID: 36102060 PMCID: PMC9471990 DOI: 10.1098/rsob.210356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Fibroblasts are widely distributed cells found in most tissues and upon tissue injury, they are able to differentiate into myofibroblasts, which express abundant extracellular matrix (ECM) proteins. Overexpression and unordered organization of ECM proteins cause tissue fibrosis in damaged tissue. Fibroblast growth factor (FGF) family proteins are well known to promote angiogenesis and tissue repair, but their activities in fibroblast differentiation and fibrosis have not been systematically reviewed. Here we summarize the effects of FGFs in fibroblast to myofibroblast differentiation and ECM protein expression and discuss the underlying potential regulatory mechanisms, to provide a basis for the clinical application of recombinant FGF protein drugs in treatment of tissue damage.
Collapse
Affiliation(s)
- Changye Sun
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Xiangqin Tian
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Yangyang Jia
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, People's Republic of China
| | - Mingming Yang
- Department of Cardiology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, People's Republic of China
| | - Yong Li
- Department of Biochemistry, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - David G Fernig
- Department of Biochemistry, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| |
Collapse
|
16
|
Li R, Jia Y, Kong X, Nie Y, Deng Y, Liu Y. Novel drug delivery systems and disease models for pulmonary fibrosis. J Control Release 2022; 348:95-114. [PMID: 35636615 DOI: 10.1016/j.jconrel.2022.05.039] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/17/2022] [Accepted: 05/21/2022] [Indexed: 12/19/2022]
Abstract
Pulmonary fibrosis (PF) is a serious and progressive lung disease which is possibly life-threatening. It causes lung scarring and affects lung functions including epithelial cell injury, massive recruitment of immune cells and abnormal accumulation of extracellular matrix (ECM). There is currently no cure for PF. Treatment for PF is aimed at slowing the course of the disease and relieving symptoms. Pirfenidone (PFD) and nintedanib (NDNB) are currently the only two FDA-approved oral medicines to slow down the progress of idiopathic pulmonary fibrosis, a specific type of PF. Novel drug delivery systems and therapies have been developed to improve the prognosis of the disease, as well as reduce or minimize the toxicities during drug treatment. The drug delivery routes for these therapies are various including oral, intravenous, nasal, inhalant, intratracheal and transdermal; although this is dependent on specific treatment mechanisms. In addition, researchers have also expanded current animal models that could not fully restore the clinicopathology, and developed a series of in vitro models such as organoids to study the pathogenesis and treatment of PF. This review describes recent advances on pathogenesis exploration, classifies and specifies the progress of drug delivery systems by their delivery routes, as well as an overview on the in vitro and in vivo models for PF research.
Collapse
Affiliation(s)
- Rui Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yizhen Jia
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Xiaohan Kong
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yichu Nie
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan 528000, China
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yang Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China; School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
17
|
Luengen AE, Cheremkhina M, Gonzalez-Rubio J, Weckauf J, Kniebs C, Uebner H, Buhl EM, Taube C, Cornelissen CG, Schmitz-Rode T, Jockenhoevel S, Thiebes AL. Bone Marrow Derived Mesenchymal Stromal Cells Promote Vascularization and Ciliation in Airway Mucosa Tri-Culture Models in Vitro. Front Bioeng Biotechnol 2022; 10:872275. [PMID: 35782511 PMCID: PMC9247357 DOI: 10.3389/fbioe.2022.872275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Patients suffering from irresectable tracheal stenosis often face limited treatment options associated with low quality of life. To date, an optimal tracheal replacement strategy does not exist. A tissue-engineered tracheal substitute promises to overcome limitations such as implant vascularization, functional mucociliary clearance and mechanical stability. In order to advance a tracheal mucosa model recently developed by our group, we examined different supporting cell types in fibrin-based tri-culture with primary human umbilical vein endothelial cells (HUVEC) and primary human respiratory epithelial cells (HRE). Bone marrow-derived mesenchymal stromal cells (BM-MSC), adipose-derived mesenchymal stromal cells (ASC) and human nasal fibroblasts (HNF) were compared regarding their ability to promote mucociliary differentiation and vascularization in vitro. Three-dimensional co-cultures of the supporting cell types with either HRE or HUVEC were used as controls. Mucociliary differentiation and formation of vascular-like structures were analyzed by scanning electron microscopy (SEM), periodic acid Schiff’s reaction (PAS reaction), two-photon laser scanning microscopy (TPLSM) and immunohistochemistry. Cytokine levels of vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), interleukin-6 (IL6), interleukin-8 (IL8), angiopoietin 1, angiopoietin 2, fibroblast growth factor basic (FGF-b) and placenta growth factor (PIGF) in media supernatant were investigated using LEGENDplex™ bead-based immunoassay. Epithelial morphology of tri-cultures with BM-MSC most closely resembled native respiratory epithelium with respect to ciliation, mucus production as well as expression and localization of epithelial cell markers pan-cytokeratin, claudin-1, α-tubulin and mucin5AC. This was followed by tri-cultures with HNF, while ASC-supported tri-cultures lacked mucociliary differentiation. For all supporting cell types, a reduced ciliation was observed in tri-cultures compared to the corresponding co-cultures. Although formation of vascular-like structures was confirmed in all cultures, vascular networks in BM-MSC-tri-cultures were found to be more branched and extended. Concentrations of pro-angiogenic and inflammatory cytokines, in particular VEGF and angiopoietin 2, revealed to be reduced in tri-cultures compared to co-cultures. With these results, our study provides an important step towards a vascularized and ciliated tissue-engineered tracheal replacement. Additionally, our tri-culture model may in the future contribute to an improved understanding of cell-cell interactions in diseases associated with impaired mucosal function.
Collapse
Affiliation(s)
- Anja E. Luengen
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, Netherlands
| | - Maria Cheremkhina
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, Netherlands
| | - Julian Gonzalez-Rubio
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, Netherlands
| | - Jan Weckauf
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, Netherlands
| | - Caroline Kniebs
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, Netherlands
| | - Hendrik Uebner
- Department of Pulmonary Medicine, University Medical Center Essen—Ruhrlandklinik, Essen, Germany
| | - E. Miriam Buhl
- Institute of Pathology, Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Medical Center Essen—Ruhrlandklinik, Essen, Germany
| | - Christian G. Cornelissen
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Clinic for Pneumology and Internal Intensive Care Medicine (Medical Clinic V), RWTH Aachen University Hospital, Aachen, Germany
| | - Thomas Schmitz-Rode
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Stefan Jockenhoevel
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, Netherlands
- *Correspondence: Stefan Jockenhoevel, ; Anja Lena Thiebes,
| | - Anja Lena Thiebes
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, Geleen, Netherlands
- *Correspondence: Stefan Jockenhoevel, ; Anja Lena Thiebes,
| |
Collapse
|
18
|
Hiller BE, Yin Y, Perng YC, de Araujo Castro Í, Fox LE, Locke MC, Monte KJ, López CB, Ornitz DM, Lenschow DJ. Fibroblast growth factor-9 expression in airway epithelial cells amplifies the type I interferon response and alters influenza A virus pathogenesis. PLoS Pathog 2022; 18:e1010228. [PMID: 35675358 PMCID: PMC9212157 DOI: 10.1371/journal.ppat.1010228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 06/21/2022] [Accepted: 05/16/2022] [Indexed: 11/19/2022] Open
Abstract
Influenza A virus (IAV) preferentially infects conducting airway and alveolar epithelial cells in the lung. The outcome of these infections is impacted by the host response, including the production of various cytokines, chemokines, and growth factors. Fibroblast growth factor-9 (FGF9) is required for lung development, can display antiviral activity in vitro, and is upregulated in asymptomatic patients during early IAV infection. We therefore hypothesized that FGF9 would protect the lungs from respiratory virus infection and evaluated IAV pathogenesis in mice that overexpress FGF9 in club cells in the conducting airway epithelium (FGF9-OE mice). However, we found that FGF9-OE mice were highly susceptible to IAV and Sendai virus infection compared to control mice. FGF9-OE mice displayed elevated and persistent viral loads, increased expression of cytokines and chemokines, and increased numbers of infiltrating immune cells as early as 1 day post-infection (dpi). Gene expression analysis showed an elevated type I interferon (IFN) signature in the conducting airway epithelium and analysis of IAV tropism uncovered a dramatic shift in infection from the conducting airway epithelium to the alveolar epithelium in FGF9-OE lungs. These results demonstrate that FGF9 signaling primes the conducting airway epithelium to rapidly induce a localized IFN and proinflammatory cytokine response during viral infection. Although this response protects the airway epithelial cells from IAV infection, it allows for early and enhanced infection of the alveolar epithelium, ultimately leading to increased morbidity and mortality. Our study illuminates a novel role for FGF9 in regulating respiratory virus infection and pathogenesis. Influenza viruses are respiratory viruses that cause significant morbidity and mortality worldwide. In the lungs, influenza A virus primarily infects epithelial cells that line the conducting airways and alveoli. Fibroblast growth factor-9 (FGF9) is a growth factor that has been shown to have antiviral activity and is upregulated during early IAV infection in asymptomatic patients, leading us to hypothesize that FGF9 would protect the lung epithelium from IAV infection. However, mice that express and secrete FGF9 from club cells in the conducting airway had more severe respiratory virus infection and a hyperactive inflammatory immune response as early as 1 day post-infection. Analysis of the FGF9-expressing airway epithelial cells found an elevated antiviral and inflammatory interferon signature, which protected these cells from severe IAV infection. However, heightened infection of alveolar cells resulted in excessive inflammation in the alveoli, resulting in more severe disease and death. Our study identifies a novel antiviral and inflammatory role for FGFs in the lung airway epithelium and confirms that early and robust IAV infection of alveolar cells results in more severe disease.
Collapse
Affiliation(s)
- Bradley E Hiller
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Yongjun Yin
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, Unites States of America
| | - Yi-Chieh Perng
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ítalo de Araujo Castro
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Center for Women Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, Unites States of America
| | - Lindsey E Fox
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Marissa C Locke
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kristen J Monte
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Carolina B López
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Center for Women Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, Unites States of America
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, Unites States of America
| | - Deborah J Lenschow
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
19
|
Dolivo DM. Anti-fibrotic effects of pharmacologic FGF-2: a review of recent literature. J Mol Med (Berl) 2022; 100:847-860. [PMID: 35484303 DOI: 10.1007/s00109-022-02194-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/09/2022] [Accepted: 03/28/2022] [Indexed: 02/06/2023]
Abstract
Fibrosis is a process of pathological tissue repair that replaces damaged, formerly functional tissue with a non-functional, collagen-rich scar. Complications of fibrotic pathologies, which can arise in numerous organs and from numerous conditions, result in nearly half of deaths in the developed world. Despite this, therapies that target fibrosis at its mechanistic roots are still notably lacking. The ubiquity of the occurrence of fibrosis in myriad organs emphasizes the fact that there are shared mechanisms underlying fibrotic conditions, which may serve as common therapeutic targets for multiple fibrotic diseases of varied organs. Thus, study of the basic science of fibrosis and of anti-fibrotic modalities is critical to therapeutic development and may have potential to translate across organs and disease states. Fibroblast growth factor 2 (FGF-2) is a broadly studied member of the fibroblast growth factors, a family of multipotent cytokines implicated in diverse cellular and tissue processes, which has previously been recognized for its anti-fibrotic potential. However, the mechanisms underlying this potential are not fully understood, nor is the potential for its use to ameliorate fibrosis in diverse pathologies and tissues. Presented here is a review of recent literature that sheds further light on these questions, with the hopes of inspiring further research into the mechanisms underlying the anti-fibrotic activities of FGF-2, as well as the disease conditions for which pharmacologic FGF-2 might be a useful option in the future.
Collapse
|
20
|
Bronchoalveolar-Lavage-Derived Fibroblast Cell Line (B-LSDM7) as a New Protocol for Investigating the Mechanisms of Idiopathic Pulmonary Fibrosis. Cells 2022; 11:cells11091441. [PMID: 35563747 PMCID: PMC9103910 DOI: 10.3390/cells11091441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 12/27/2022] Open
Abstract
Background: The use of BAL to study ILDs has improved our understanding of IPF pathogenesis. BAL fluid is routinely collected and can be considered a clinical and research tool. The procedure is well tolerated and minimally invasive. No specific cell lines from BAL or immortalized cell lines from IPF patients are available commercially. A method to quickly isolate and characterize fibroblasts from BAL is an unmet research need. Materials and methods: Here we describe a new protocol by which we isolated a cell line from IPF. The cell line was expanded in vitro and characterized phenotypically, morphologically and functionally. Results: This culture showed highly filamentous cells with an evident central nucleus. From the phenotypic point of view, this cell line displays fibroblast/myofibroblast-like features including expression of alpha-SMA, vimentin, collagen type-1 and fibronectin. The results showed high expression of ROS in these cells. Oxidative stress invariably promotes extracellular matrix expression in lung diseases directly or through over-production of pro-fibrotic growth factors. Conclusions: Our protocol makes it possible to obtain fibroblasts BAL that is a routine non-invasive method that offers the possibility of having a large sample of patients. Standardized culture methods are important for a reliable model for testing molecules and eventual novel development therapeutic targets.
Collapse
|
21
|
Guidi R, Xu D, Choy DF, Ramalingam TR, Lee WP, Modrusan Z, Liang Y, Marsters S, Ashkenazi A, Huynh A, Mills J, Flanagan S, Hambro S, Nunez V, Leong L, Cook A, Tran TH, Austin CD, Cao Y, Clarke C, Panettieri RA, Koziol-White C, Jester WF, Wang F, Wilson MS. Steroid-induced fibroblast growth factors drive an epithelial-mesenchymal inflammatory axis in severe asthma. Sci Transl Med 2022; 14:eabl8146. [PMID: 35442706 PMCID: PMC10301263 DOI: 10.1126/scitranslmed.abl8146] [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] [Indexed: 11/02/2022]
Abstract
Asthma and inflammatory airway diseases restrict airflow in the lung, compromising gas exchange and lung function. Inhaled corticosteroids (ICSs) can reduce inflammation, control symptoms, and improve lung function; however, a growing number of patients with severe asthma do not benefit from ICS. Using bronchial airway epithelial brushings from patients with severe asthma or primary human cells, we delineated a corticosteroid-driven fibroblast growth factor (FGF)-dependent inflammatory axis, with FGF-responsive fibroblasts promoting downstream granulocyte colony-stimulating factor (G-CSF) production, hyaluronan secretion, and neutrophilic inflammation. Allergen challenge studies in mice demonstrate that the ICS, fluticasone propionate, inhibited type 2-driven eosinophilia but induced a concomitant increase in FGFs, G-CSF, hyaluronan, and neutrophil infiltration. We developed a model of steroid-induced neutrophilic inflammation mediated, in part, by induction of an FGF-dependent epithelial-mesenchymal axis, which may explain why some individuals do not benefit from ICS. In further proof-of-concept experiments, we found that combination therapy with pan-FGF receptor inhibitors and corticosteroids prevented both eosinophilic and steroid-induced neutrophilic inflammation. Together, these results establish FGFs as therapeutic targets for severe asthma patients who do not benefit from ICS.
Collapse
Affiliation(s)
- Riccardo Guidi
- Immunology Discovery, Genentech, South San Francisco, CA 94080, USA
| | - Daqi Xu
- Immunology Discovery, Genentech, South San Francisco, CA 94080, USA
| | - David F. Choy
- Biomarker Discovery OMNI, Genentech, South San Francisco, CA 94080, USA
| | | | - Wyne P. Lee
- Translational Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Zora Modrusan
- Next Generation Sequencing (NGS), Genentech, South San Francisco, CA 94080, USA
| | - Yuxin Liang
- Next Generation Sequencing (NGS), Genentech, South San Francisco, CA 94080, USA
| | - Scot Marsters
- Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Avi Ashkenazi
- Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Alison Huynh
- Necropsy, Genentech, South San Francisco, CA 94080, USA
| | - Jessica Mills
- Necropsy, Genentech, South San Francisco, CA 94080, USA
| | - Sean Flanagan
- Necropsy, Genentech, South San Francisco, CA 94080, USA
| | | | - Victor Nunez
- Necropsy, Genentech, South San Francisco, CA 94080, USA
| | - Laurie Leong
- Pathology, Genentech, South San Francisco, CA 94080, USA
| | - Ashley Cook
- Pathology, Genentech, South San Francisco, CA 94080, USA
| | | | - Cary D. Austin
- Pathology, Genentech, South San Francisco, CA 94080, USA
| | - Yi Cao
- OMNI Bioinformatics, Genentech, South San Francisco, CA 94080, USA
| | - Christine Clarke
- OMNI Bioinformatics, Genentech, South San Francisco, CA 94080, USA
| | - Reynold A. Panettieri
- Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Cynthia Koziol-White
- Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - William F. Jester
- Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Fen Wang
- Center for Cancer Biology and Nutrition, Texas A&M University, Houston, TX 77030, USA
| | - Mark S. Wilson
- Immunology Discovery, Genentech, South San Francisco, CA 94080, USA
| |
Collapse
|
22
|
Chen K, Rao Z, Dong S, Chen Y, Wang X, Luo Y, Gong F, Li X. Roles of the fibroblast growth factor signal transduction system in tissue injury repair. BURNS & TRAUMA 2022; 10:tkac005. [PMID: 35350443 PMCID: PMC8946634 DOI: 10.1093/burnst/tkac005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/13/2021] [Indexed: 12/13/2022]
Abstract
Following injury, tissue autonomously initiates a complex repair process, resulting in either partial recovery or regeneration of tissue architecture and function in most organisms. Both the repair and regeneration processes are highly coordinated by a hierarchy of interplay among signal transduction pathways initiated by different growth factors, cytokines and other signaling molecules under normal conditions. However, under chronic traumatic or pathological conditions, the reparative or regenerative process of most tissues in different organs can lose control to different extents, leading to random, incomplete or even flawed cell and tissue reconstitution and thus often partial restoration of the original structure and function, accompanied by the development of fibrosis, scarring or even pathogenesis that could cause organ failure and death of the organism. Ample evidence suggests that the various combinatorial fibroblast growth factor (FGF) and receptor signal transduction systems play prominent roles in injury repair and the remodeling of adult tissues in addition to embryonic development and regulation of metabolic homeostasis. In this review, we attempt to provide a brief update on our current understanding of the roles, the underlying mechanisms and clinical application of FGFs in tissue injury repair.
Collapse
Affiliation(s)
| | | | - Siyang Dong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Department of breast surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yajing Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xulan Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yongde Luo
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| | - Fanghua Gong
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| | - Xiaokun Li
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| |
Collapse
|
23
|
Farooq M, Khan AW, Kim MS, Choi S. The Role of Fibroblast Growth Factor (FGF) Signaling in Tissue Repair and Regeneration. Cells 2021; 10:cells10113242. [PMID: 34831463 PMCID: PMC8622657 DOI: 10.3390/cells10113242] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/10/2021] [Accepted: 11/16/2021] [Indexed: 02/06/2023] Open
Abstract
Fibroblast growth factors (FGFs) are a large family of secretory molecules that act through tyrosine kinase receptors known as FGF receptors. They play crucial roles in a wide variety of cellular functions, including cell proliferation, survival, metabolism, morphogenesis, and differentiation, as well as in tissue repair and regeneration. The signaling pathways regulated by FGFs include RAS/mitogen-activated protein kinase (MAPK), phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)–protein kinase B (AKT), phospholipase C gamma (PLCγ), and signal transducer and activator of transcription (STAT). To date, 22 FGFs have been discovered, involved in different functions in the body. Several FGFs directly or indirectly interfere with repair during tissue regeneration, in addition to their critical functions in the maintenance of pluripotency and dedifferentiation of stem cells. In this review, we summarize the roles of FGFs in diverse cellular processes and shed light on the importance of FGF signaling in mechanisms of tissue repair and regeneration.
Collapse
Affiliation(s)
- Mariya Farooq
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
| | - Abdul Waheed Khan
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Korea
- Correspondence:
| |
Collapse
|
24
|
D’Agnillo F, Walters KA, Xiao Y, Sheng ZM, Scherler K, Park J, Gygli S, Rosas LA, Sadtler K, Kalish H, Blatti CA, Zhu R, Gatzke L, Bushell C, Memoli MJ, O’Day SJ, Fischer TD, Hammond TC, Lee RC, Cash JC, Powers ME, O’Keefe GE, Butnor KJ, Rapkiewicz AV, Travis WD, Layne SP, Kash JC, Taubenberger JK. Lung epithelial and endothelial damage, loss of tissue repair, inhibition of fibrinolysis, and cellular senescence in fatal COVID-19. Sci Transl Med 2021; 13:eabj7790. [PMID: 34648357 PMCID: PMC11000440 DOI: 10.1126/scitranslmed.abj7790] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is characterized by respiratory distress, multiorgan dysfunction, and, in some cases, death. The pathological mechanisms underlying COVID-19 respiratory distress and the interplay with aggravating risk factors have not been fully defined. Lung autopsy samples from 18 patients with fatal COVID-19, with symptom onset-to-death times ranging from 3 to 47 days, and antemortem plasma samples from 6 of these cases were evaluated using deep sequencing of SARS-CoV-2 RNA, multiplex plasma protein measurements, and pulmonary gene expression and imaging analyses. Prominent histopathological features in this case series included progressive diffuse alveolar damage with excessive thrombosis and late-onset pulmonary tissue and vascular remodeling. Acute damage at the alveolar-capillary barrier was characterized by the loss of surfactant protein expression with injury to alveolar epithelial cells, endothelial cells, respiratory epithelial basal cells, and defective tissue repair processes. Other key findings included impaired clot fibrinolysis with increased concentrations of plasma and lung plasminogen activator inhibitor-1 and modulation of cellular senescence markers, including p21 and sirtuin-1, in both lung epithelial and endothelial cells. Together, these findings further define the molecular pathological features underlying the pulmonary response to SARS-CoV-2 infection and provide important insights into signaling pathways that may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Felice D’Agnillo
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | | | - Yongli Xiao
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Zong-Mei Sheng
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Jaekeun Park
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sebastian Gygli
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Luz Angela Rosas
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kaitlyn Sadtler
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Heather Kalish
- Bioengineering and Physical Sciences Shared Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Charles A. Blatti
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ruoqing Zhu
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Lisa Gatzke
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Colleen Bushell
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Matthew J. Memoli
- Clinical Studies Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | - Raymond C. Lee
- Division of Cardiothoracic Surgery, USC Keck School of Medicine, Los Angeles, CA, USA
| | - J. Christian Cash
- Division of Cardiothoracic Surgery, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Matthew E. Powers
- Division of Cardiothoracic Surgery, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Grant E. O’Keefe
- Department of Surgery, University of Washington, Harborview Medical Center, Seattle, WA, USA
| | - Kelly J. Butnor
- Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington, VT, USA
| | - Amy V. Rapkiewicz
- Department of Pathology, New York University Long Island School of Medicine, Mineola, NY, USA
| | - William D. Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - John C. Kash
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffery K. Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
25
|
Prudovsky I. Cellular Mechanisms of FGF-Stimulated Tissue Repair. Cells 2021; 10:cells10071830. [PMID: 34360000 PMCID: PMC8304273 DOI: 10.3390/cells10071830] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/10/2023] Open
Abstract
Growth factors belonging to the FGF family play important roles in tissue and organ repair after trauma. In this review, I discuss the regulation by FGFs of the aspects of cellular behavior important for reparative processes. In particular, I focus on the FGF-dependent regulation of cell proliferation, cell stemness, de-differentiation, inflammation, angiogenesis, cell senescence, cell death, and the production of proteases. In addition, I review the available literature on the enhancement of FGF expression and secretion in damaged tissues resulting in the increased FGF supply required for tissue repair.
Collapse
Affiliation(s)
- Igor Prudovsky
- Maine Medical Center Research Institute, 81 Research Dr., Scarborough, ME 04074, USA
| |
Collapse
|
26
|
Fortier SM, Penke LR, King D, Pham TX, Ligresti G, Peters-Golden M. Myofibroblast dedifferentiation proceeds via distinct transcriptomic and phenotypic transitions. JCI Insight 2021; 6:144799. [PMID: 33561015 PMCID: PMC8026183 DOI: 10.1172/jci.insight.144799] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/04/2021] [Indexed: 12/16/2022] Open
Abstract
Myofibroblasts are the major cellular source of collagen, and their accumulation - via differentiation from fibroblasts and resistance to apoptosis - is a hallmark of tissue fibrosis. Clearance of myofibroblasts by dedifferentiation and restoration of apoptosis sensitivity has the potential to reverse fibrosis. Prostaglandin E2 (PGE2) and mitogens such as FGF2 have each been shown to dedifferentiate myofibroblasts, but - to our knowledge - the resultant cellular phenotypes have neither been comprehensively characterized or compared. Here, we show that PGE2 elicited dedifferentiation of human lung myofibroblasts via cAMP/PKA, while FGF2 utilized MEK/ERK. The 2 mediators yielded transitional cells with distinct transcriptomes, with FGF2 promoting but PGE2 inhibiting proliferation and survival. The gene expression pattern in fibroblasts isolated from the lungs of mice undergoing resolution of experimental fibrosis resembled that of myofibroblasts treated with PGE2 in vitro. We conclude that myofibroblast dedifferentiation can proceed via distinct programs exemplified by treatment with PGE2 and FGF2, with dedifferentiation occurring in vivo most closely resembling the former.
Collapse
Affiliation(s)
| | - Loka R. Penke
- Division of Pulmonary and Critical Care Medicine and
| | - Dana King
- BCRF Bioinformatics Core, University of Michigan, Ann Arbor, Michigan, USA
| | - Tho X. Pham
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Giovanni Ligresti
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | | |
Collapse
|
27
|
Nickol ME, Lyle SM, Dennehy B, Kindrachuk J. Dysregulated Host Responses Underlie 2009 Pandemic Influenza-Methicillin Resistant Staphylococcus aureus Coinfection Pathogenesis at the Alveolar-Capillary Barrier. Cells 2020; 9:E2472. [PMID: 33202895 PMCID: PMC7696554 DOI: 10.3390/cells9112472] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 02/04/2023] Open
Abstract
Influenza viruses are a continual public health concern resulting in 3-5 million severe infections annually despite intense vaccination campaigns and messaging. Secondary bacterial infections, including Staphylococcus aureus, result in increased morbidity and mortality during seasonal epidemics and pandemics. While coinfections can result in deleterious pathologic consequences, including alveolar-capillary barrier disruption, the underlying mechanisms are poorly understood. We have characterized host- and pathogen-centric mechanisms contributing to influenza-bacterial coinfections in a primary cell coculture model of the alveolar-capillary barrier. Using 2009 pandemic influenza (pH1N1) and methicillin-resistant S. aureus (MRSA), we demonstrate that coinfection resulted in dysregulated barrier function. Preinfection with pH1N1 resulted in modulation of adhesion- and invasion-associated MRSA virulence factors during lag phase bacterial replication. Host response modulation in coinfected alveolar epithelial cells were primarily related to TLR- and inflammatory response-mediated cell signaling events. While less extensive in cocultured endothelial cells, coinfection resulted in changes to cellular stress response- and TLR-related signaling events. Analysis of cytokine expression suggested that cytokine secretion might play an important role in coinfection pathogenesis. Taken together, we demonstrate that coinfection pathogenesis is related to complex host- and pathogen-mediated events impacting both epithelial and endothelial cell regulation at the alveolar-capillary barrier.
Collapse
Affiliation(s)
- Michaela E. Nickol
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (M.E.N.); (S.M.L.); (B.D.)
| | - Sarah M. Lyle
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (M.E.N.); (S.M.L.); (B.D.)
| | - Brendan Dennehy
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (M.E.N.); (S.M.L.); (B.D.)
| | - Jason Kindrachuk
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (M.E.N.); (S.M.L.); (B.D.)
- Vaccine and Infectious Disease Organization-International Vaccine Centre, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| |
Collapse
|
28
|
El-Baz LMF, Shoukry NM, Hafez HS, Guzy RD, Salem ML. Fibroblast Growth Factor 2 Augments Transforming Growth Factor Beta 1 Induced Epithelial-mesenchymal Transition in Lung Cell Culture Model. IRANIAN JOURNAL OF ALLERGY, ASTHMA, AND IMMUNOLOGY 2020; 19:348-361. [PMID: 33463102 PMCID: PMC8366022 DOI: 10.18502/ijaai.v19i4.4110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 05/27/2020] [Indexed: 02/05/2023]
Abstract
Impaired lung epithelial cell regeneration following injury may contribute to the development of pulmonary fibrosis. Epithelial-mesenchymal transition (EMT) is a critical event in embryonic development, wound healing following injury, and even cancer progression. Previous studies have shown that the combination of transforming growth factor beta-1 (TGFβ1) and fibroblast growth factor 2 (FGF2) induces EMT during cancer metastasis. However, this synergy remains to be elucidated in inducing EMT associated with wound healing after injury. We set out this study to determine the effect of fibroblast growth factor 2 (FGF2) on TGFβ1-induced EMT in the human lung epithelium. BEAS-2B and A549 cells were treated with TGFβ1, FGF2, or both. EMT phenotype was investigated morphologically and by measuring mRNA expression levels; using quantitative real-time PCR. E-cadherin expression was assayed by western blot and immunofluorescence staining. Cell migration was confirmed using a wound-healing assay. TGFβ1 induced a morphological change and a significant increase in cell migration of BEAS-2B cells. TGFβ1 significantly reduced E-cadherin (CDH1) mRNA expression and markedly induced expression of N-cadherin (CDH2), tenascin C (TNC), fibronectin (FN), actin alpha 2 (ACTA2), and collagen I (COL1A1). While FGF2 alone did not significantly alter EMT gene expression, it enhanced TGFβ1-induced suppression of CDH1 and upregulation of ACTA2, but not TNC, FN, and CDH2. FGF2 significantly inhibited TGFβ1-induced COL1A1 expression. Furthermore, FGF2 maintained TGFβ1-induced morphologic changes and increased the migration of TGFβ1-treated cells. This study suggests a synergistic effect between TGFβ1 and FGF2 in inducing EMT in lung epithelial cells, which may play an important role in wound healing and tissue repair after injury.
Collapse
Affiliation(s)
- Lamis M F El-Baz
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois, USA AND Department of Zoology, Faculty of Science, Suez University, Suez, Egypt.
| | - Nahla M Shoukry
- Department of Zoology, Faculty of Science, Suez University, Suez, Egypt.
| | - Hani S Hafez
- Department of Zoology, Faculty of Science, Suez University, Suez, Egypt.
| | - Robert D Guzy
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois, USA.
| | - Mohamed Labib Salem
- Department of Immunology and Biotechnology, Faculty of Science, Center of Excellence in Cancer Research, Tanta University, Tanta, Egypt.
| |
Collapse
|
29
|
Shimbori C, El Agha E. Good Things Come in 2s: Type 2 Alveolar Epithelial Cells and Fibroblast Growth Factor Receptor 2. Am J Respir Cell Mol Biol 2020; 62:543-545. [PMID: 31940443 PMCID: PMC7193794 DOI: 10.1165/rcmb.2020-0013ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Chiko Shimbori
- Farncombe Family Digestive Health Research InstituteMcMaster UniversityHamilton, Ontario, Canadaand
| | - Elie El Agha
- Institute for Lung HealthJustus-Liebig University GiessenGiessen, Germany
| |
Collapse
|
30
|
Dorry SJ, Ansbro BO, Ornitz DM, Mutlu GM, Guzy RD. FGFR2 Is Required for AEC2 Homeostasis and Survival after Bleomycin-induced Lung Injury. Am J Respir Cell Mol Biol 2020; 62:608-621. [PMID: 31860803 PMCID: PMC7193788 DOI: 10.1165/rcmb.2019-0079oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 12/20/2019] [Indexed: 12/28/2022] Open
Abstract
Alveolar epithelial cell (AEC) injury is central to the pathogenesis of pulmonary fibrosis. Epithelial FGF (fibroblast growth factor) signaling is essential for recovery from hyperoxia- and influenza-induced lung injury, and treatment with FGFs is protective in experimental lung injury. The cell types involved in the protective effect of FGFs are not known. We hypothesized that FGF signaling in type II AECs (AEC2s) is critical in bleomycin-induced lung injury and fibrosis. To test this hypothesis, we generated mice with tamoxifen-inducible deletion of FGFR1-3 (fibroblast growth factor receptors 1, 2, and 3) in surfactant protein C-positive (SPC+) AEC2s (SPC triple conditional knockout [SPC-TCKO]). In the absence of injury, SPC-TCKO mice had fewer AEC2s, decreased Sftpc (surfactant protein C gene) expression, increased alveolar diameter, and increased collagen deposition. After intratracheal bleomycin administration, SPC-TCKO mice had increased mortality, lung edema, and BAL total protein, and flow cytometry and immunofluorescence revealed a loss of AEC2s. To reduce mortality of SPC-TCKO mice to less than 50%, a 25-fold dose reduction of bleomycin was required. Surviving bleomycin-injured SPC-TCKO mice had increased collagen deposition, fibrosis, and ACTA2 expression and decreased epithelial gene expression. Inducible inactivation of individual Fgfr2 or Fgfr3 revealed that Fgfr2, but not Fgfr3, was responsible for the increased mortality and lung injury after bleomycin administration. In conclusion, AEC2-specific FGFR2 is critical for survival in response to bleomycin-induced lung injury. These data also suggest that a population of SPC+ AEC2s require FGFR2 signaling for maintenance in the adult lung. Preventing epithelial FGFR inhibition and/or activating FGFRs in alveolar epithelium may therefore represent a novel approach to treating lung injury and reducing fibrosis.
Collapse
Affiliation(s)
- Samuel J. Dorry
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois; and
| | - Brandon O. Ansbro
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois; and
| | - David M. Ornitz
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, Missouri
| | - Gökhan M. Mutlu
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois; and
| | - Robert D. Guzy
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois; and
| |
Collapse
|
31
|
Luengen AE, Kniebs C, Buhl EM, Cornelissen CG, Schmitz-Rode T, Jockenhoevel S, Thiebes AL. Choosing the Right Differentiation Medium to Develop Mucociliary Phenotype of Primary Nasal Epithelial Cells In Vitro. Sci Rep 2020; 10:6963. [PMID: 32332878 PMCID: PMC7181704 DOI: 10.1038/s41598-020-63922-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/06/2020] [Indexed: 11/08/2022] Open
Abstract
In vitro differentiation of airway epithelium is of interest for respiratory tissue engineering and studying airway diseases. Both applications benefit from the use of primary cells to maintain a mucociliated phenotype and thus physiological functionality. Complex differentiation procedures often lack standardization and reproducibility. To alleviate these shortfalls, we compared differentiation behavior of human nasal epithelial cells in four differentiation media. Cells were differentiated at the air-liquid interface (ALI) on collagen-coated inserts. Mucociliary differentiation status after five weeks was analyzed by electron microscopy, histology and immunohistochemistry. The amount of ciliation was estimated and growth factor concentrations were evaluated using ELISA. We found that retinoic-acid-supplemented mixture of DMEM and Airway Epithelial Cell Growth Medium gave most promising results to obtain ciliated and mucus producing nasal epithelium in vitro. We discovered the balance between retinoic acid (RA), vascular endothelial growth factor (VEGF), epidermal growth factor (EGF) and fibroblast growth factor β (FGF-β) to be relevant for differentiation. We could show that low VEGF, EGF and FGF-β concentrations in medium correspond to absent ciliation in specific donors. Therefore, our results may in future facilitate donor selection and non-invasive monitoring of ALI cultures and by this contribute to improved standardization of epithelial in vitro culture.
Collapse
Affiliation(s)
- Anja E Luengen
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany.
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, 6167 RD, Geleen, The Netherlands.
| | - Caroline Kniebs
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, 6167 RD, Geleen, The Netherlands
| | - Eva Miriam Buhl
- Institute of Pathology, Electron Microscopy Facility, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Christian G Cornelissen
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany
- Clinic for Pneumology and Internal Intensive Care Medicine (Medical Clinic V), RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Thomas Schmitz-Rode
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany
| | - Stefan Jockenhoevel
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany.
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, 6167 RD, Geleen, The Netherlands.
| | - Anja Lena Thiebes
- Department of Biohybrid and Medical Textiles (BioTex), AME - Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Brightlands Chemelot Campus, 6167 RD, Geleen, The Netherlands
| |
Collapse
|
32
|
Tan Y, Qiao Y, Chen Z, Liu J, Guo Y, Tran T, Tan KS, Wang DY, Yan Y. FGF2, an Immunomodulatory Factor in Asthma and Chronic Obstructive Pulmonary Disease (COPD). Front Cell Dev Biol 2020; 8:223. [PMID: 32300593 PMCID: PMC7142218 DOI: 10.3389/fcell.2020.00223] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/16/2020] [Indexed: 12/14/2022] Open
Abstract
The fibroblast growth factor 2 (FGF2) is a potent mitogenic factor belonging to the FGF family. It plays a role in airway remodeling associated with chronic inflammatory airway diseases, including asthma and chronic obstructive pulmonary disease (COPD). Recently, research interest has been raised in the immunomodulatory function of FGF2 in asthma and COPD, through its involvement in not only the regulation of inflammatory cells but also its participation as a mediator between immune cells and airway structural cells. Herein, this review provides the current knowledge on the biology of FGF2, its expression pattern in asthma and COPD patients, and its role as an immunomodulatory factor. The potential that FGF2 is involved in regulating inflammation indicates that FGF2 could be a therapeutic target for chronic inflammatory diseases.
Collapse
Affiliation(s)
- Yuanyang Tan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | | | - Zhuanggui Chen
- Department of Pediatrics, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing Liu
- Department of Respiratory Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Yanrong Guo
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Thai Tran
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kai Sen Tan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, University Health System, National University of Singapore, Singapore, Singapore
| | - De-Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, University Health System, National University of Singapore, Singapore, Singapore
| | - Yan Yan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.,Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
33
|
Koyama K, Goto H, Morizumi S, Kagawa K, Nishimura H, Sato S, Kawano H, Toyoda Y, Ogawa H, Homma S, Nishioka Y. The Tyrosine Kinase Inhibitor TAS-115 Attenuates Bleomycin-induced Lung Fibrosis in Mice. Am J Respir Cell Mol Biol 2019; 60:478-487. [PMID: 30540913 DOI: 10.1165/rcmb.2018-0098oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The signaling pathways of growth factors, including platelet-derived growth factor, can be considered specific targets for overcoming the poor prognosis of idiopathic pulmonary fibrosis. Nintedanib, the recently approved multiple kinase inhibitor, has shown promising antifibrotic effects in patients with idiopathic pulmonary fibrosis; however, its efficacy is still limited, and in some cases, treatment discontinuation is necessary owing to toxicities such as gastrointestinal disorders. Therefore, more effective agents with less toxicity are still needed. TAS-115 is a novel multiple tyrosine kinase inhibitor that preferably targets platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor receptor, and c-FMS in addition to other molecules. In this study, we evaluated the antifibrotic effect of TAS-115 on pulmonary fibrosis in vitro and in vivo. TAS-115 inhibited the phosphorylation of PDGFR on human lung fibroblast cell line MRC-5 cells and suppressed their platelet-derived growth factor-induced proliferation and migration. Furthermore, TAS-115 inhibited the phosphorylation of c-FMS, a receptor of macrophage colony-stimulating factor, in murine bone marrow-derived macrophages and decreased the production of CCL2, another key molecule for inducing pulmonary fibrosis, under the stimulation of macrophage colony-stimulating factor. Importantly, the inhibitory effects of TAS-115 on both PDGFR and c-FMS were 3- to 10-fold higher than those of nintedanib. In a mouse model of bleomycin-induced pulmonary fibrosis, TAS-115 significantly inhibited the development of pulmonary fibrosis and the collagen deposition in bleomycin-treated lungs. These data suggest that strong inhibition of PDGFR and c-FMS by TAS-115 may be a promising strategy for overcoming the intractable pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Kazuya Koyama
- 1 Department of Respiratory Medicine and Rheumatology and.,2 Department of Respiratory Medicine, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Hisatsugu Goto
- 1 Department of Respiratory Medicine and Rheumatology and
| | - Shun Morizumi
- 1 Department of Respiratory Medicine and Rheumatology and
| | - Kozo Kagawa
- 1 Department of Respiratory Medicine and Rheumatology and
| | | | - Seidai Sato
- 1 Department of Respiratory Medicine and Rheumatology and
| | - Hiroshi Kawano
- 1 Department of Respiratory Medicine and Rheumatology and
| | - Yuko Toyoda
- 1 Department of Respiratory Medicine and Rheumatology and
| | - Hirohisa Ogawa
- 3 Department of Molecular and Environmental Pathology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan; and
| | - Sakae Homma
- 2 Department of Respiratory Medicine, Toho University Graduate School of Medicine, Tokyo, Japan
| | | |
Collapse
|
34
|
Li Y, Guo XB, Wang JS, Wang HC, Li LP. Function of fibroblast growth factor 2 in gastric cancer occurrence and prognosis. Mol Med Rep 2019; 21:575-582. [PMID: 31789423 PMCID: PMC6947937 DOI: 10.3892/mmr.2019.10850] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 09/18/2019] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to explore the role of fibroblast growth factor 2 (FGF2) in the development and prognosis of gastric cancer (GC). The relationship between FGF2 mRNA expression levels and the clinical characteristics of GC was investigated using microarray data from four GC cohorts involving 726 patients obtained from the Gene Expression Omnibus. The results of the present study indicated that FGF2 expression levels were an independent factor affecting the prognosis of GC. The primary functions of FGF2 were related to cell adhesion and angiogenesis, and patients with high levels of FGF2 expression had poorer TNM staging and prognosis; these differences were statistically significant. In terms of immune infiltration, a higher extent of M2 macrophage intrusion was observed in patients with higher levels of FGF2. However, the degree of infiltration by dendritic and CD4+ T cells was lower, and this difference was statistically significant. Multivariate Cox proportional hazards model analysis revealed that age, TNM staging and FGF2 expression levels were independent prognostic factors for GC. In summary, FGF2 expression was demonstrated to be an independent prognostic factor in GC, and higher levels of FGF2 may promote the progression of this malignancy.
Collapse
Affiliation(s)
- Yang Li
- Gastrointestinal Department of Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250000, P.R. China
| | - Xiao-Bo Guo
- Gastrointestinal Department of Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250000, P.R. China
| | - Jin Shen Wang
- Gastrointestinal Department of Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250000, P.R. China
| | - Hong-Chang Wang
- Gastrointestinal Department of Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250000, P.R. China
| | - Le-Ping Li
- Gastrointestinal Department of Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250000, P.R. China
| |
Collapse
|
35
|
Logan SM, Storey KB. Angiogenic signaling in the lungs of a metabolically suppressed hibernating mammal ( Ictidomys tridecemlineatus). PeerJ 2019; 7:e8116. [PMID: 31763078 PMCID: PMC6870509 DOI: 10.7717/peerj.8116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/29/2019] [Indexed: 01/18/2023] Open
Abstract
To conserve energy in times of limited resource availability, particularly during cold winters, hibernators suppress even the most basic of physiologic processes. Breathing rates decrease from 40 breaths/minute to less than 1 breath/min as they decrease body temperature from 37 °C to ambient. Nevertheless, after months of hibernation, these incredible mammals emerge from torpor unscathed. This study was conducted to better understand the protective and possibly anti-inflammatory adaptations that hibernator lungs may use to prevent damage associated with entering and emerging from natural torpor. We postulated that the differential protein expression of soluble protein receptors (decoy receptors that sequester soluble ligands to inhibit signal transduction) would help identify inhibited inflammatory signaling pathways in metabolically suppressed lungs. Instead, the only two soluble receptors that responded to torpor were sVEGFR1 and sVEGFR2, two receptors whose full-length forms are bound by VEGF-A to regulate endothelial cell function and angiogenesis. Decreased sVEGFR1/2 correlated with increased total VEGFR2 protein levels. Maintained or increased levels of key γ-secretase subunits suggested that decreased sVEGFR1/2 protein levels were not due to decreased levels of intramembrane cleavage complex subunits. VEGF-A protein levels did not change, suggesting that hibernators may regulate VEGFR1/2 signaling at the level of the receptor instead of increasing relative ligand abundance. A panel of angiogenic factors used to identify biomarkers of angiogenesis showed a decrease in FGF-1 and an increase in BMP-9. Torpid lungs may use VEGF and BMP-9 signaling to balance angiogenesis and vascular stability, possibly through the activation of SMAD signaling for adaptive tissue remodeling.
Collapse
Affiliation(s)
- Samantha M. Logan
- Institute of Biochemistry, Departments of Biology and Chemistry, Carleton University, Ottawa, ON, Canada
| | - Kenneth B. Storey
- Institute of Biochemistry, Departments of Biology and Chemistry, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
36
|
Cianchetti S, Cardini C, Puxeddu I, Latorre M, Bartoli ML, Bradicich M, Dente F, Bacci E, Celi A, Paggiaro P. Distinct profile of inflammatory and remodelling biomarkers in sputum of severe asthmatic patients with or without persistent airway obstruction. World Allergy Organ J 2019; 12:100078. [PMID: 31871533 PMCID: PMC6911957 DOI: 10.1016/j.waojou.2019.100078] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 09/25/2019] [Accepted: 09/27/2019] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Both inflammatory and remodelling processes are associated with irreversible airway obstruction observed in severe asthma. Our aim was to characterize a group of severe asthmatic patients with or without persistent airway obstruction in relation to specific sputum inflammatory and remodelling biomarkers. METHODS Forty-five patients under regular high-dose inhaled corticosteroid/ß-2agonist treatment were studied, after a follow-up period of at least 2 years, with a minimum of 4 visits. Periostin, TGF-ß, RANTES, IL-8, GM-CSF, FGF-2, and cell counts were measured in induced sputum. Serum periostin was also measured. RESULTS Sputum induction was successfully performed in all but 5 patients. There were no significant differences in demographic and clinical data between patients with non-persistent obstruction (NO: FEV1/VC>88%pred.) and those with persistent obstruction (O: a not completely reversible obstruction with FEV1/VC<88%pred. at each visit before the study visit). Patients with persistent obstruction had significantly higher sputum periostin and TGF-ß concentrations than NO patients and a trend of higher serum periostin levels. GM-CSF and FGF-2 were significantly increased in NO compared to O patients. No differences between groups were found for RANTES, IL-8 and differential cell counts. Sputum periostin inversely correlated with functional parameters (prebronch. FEV1: rho = -0.36, p < 0.05; postbronch. FEV1: rho = -0.33, p = 0.05). Patients with high sputum periostin concentration (>103.3 pg/ml: median value) showed an absolute number of sputum eosinophils significantly higher than patients with low sputum periostin; this behavior was unobserved when serum periostin was considered. CONCLUSIONS Only periostin and TGF-ß identified a subgroup of severe asthmatic patients with persistent airway obstruction. Sputum periostin was also inversely associated with FEV1 and proved to be a more sensitive biomarker than serum periostin to identify severe asthmatics with higher sputum eosinophilia.
Collapse
Key Words
- Airway inflammation
- BMI, body mass index
- Biomarkers
- FEV1, forced expiratory volume in 1 s
- FGF-2, fibroblast growth factor-2
- FeNO, fraction of exhaled nitric oxide
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- ICS, inhaled corticosteroids
- IFN, interferon
- IL-8, interleukin-8
- Induced sputum
- LABA, long-acting ß-2agonist
- LTRA, leukotriene receptor antagonist
- RANTES, regulated on activation, normal T-cells expressed and secreted
- Remodelling
- Severe asthma
- TGF-ß, transforming growth factor-ß-1
- VC, vital capacity
Collapse
Affiliation(s)
- Silvana Cianchetti
- Respiratory Pathophysiology Unit, Department of Surgery, Medicine, Molecular Biology, and Critical Care, University of Pisa, Pisa, Italy
| | - Cristina Cardini
- Respiratory Pathophysiology Unit, Department of Surgery, Medicine, Molecular Biology, and Critical Care, University of Pisa, Pisa, Italy
| | - Ilaria Puxeddu
- Immunology and Allergology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Manuela Latorre
- Respiratory Pathophysiology Unit, Department of Surgery, Medicine, Molecular Biology, and Critical Care, University of Pisa, Pisa, Italy
| | - Maria Laura Bartoli
- Respiratory Pathophysiology Unit, Department of Surgery, Medicine, Molecular Biology, and Critical Care, University of Pisa, Pisa, Italy
| | - Matteo Bradicich
- Respiratory Pathophysiology Unit, Department of Surgery, Medicine, Molecular Biology, and Critical Care, University of Pisa, Pisa, Italy
| | - Federico Dente
- Respiratory Pathophysiology Unit, Department of Surgery, Medicine, Molecular Biology, and Critical Care, University of Pisa, Pisa, Italy
| | - Elena Bacci
- Respiratory Pathophysiology Unit, Department of Surgery, Medicine, Molecular Biology, and Critical Care, University of Pisa, Pisa, Italy
| | - Alessandro Celi
- Respiratory Pathophysiology Unit, Department of Surgery, Medicine, Molecular Biology, and Critical Care, University of Pisa, Pisa, Italy
| | - Pierluigi Paggiaro
- Respiratory Pathophysiology Unit, Department of Surgery, Medicine, Molecular Biology, and Critical Care, University of Pisa, Pisa, Italy
| |
Collapse
|
37
|
Wang K, Lai C, Li T, Wang C, Wang W, Ni B, Bai C, Zhang S, Han L, Gu H, Zhao Z, Duan Y, Yang X, Xing L, Zhao L, Zhou S, Xia M, Jiang C, Wang X, Yang P. Basic fibroblast growth factor protects against influenza A virus-induced acute lung injury by recruiting neutrophils. J Mol Cell Biol 2019; 10:573-585. [PMID: 29121325 DOI: 10.1093/jmcb/mjx047] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 11/02/2017] [Indexed: 12/18/2022] Open
Abstract
Influenza virus (IAV) infection is a major cause of severe respiratory illness that affects almost every country in the world. IAV infections result in respiratory illness and even acute lung injury and death, but the underlying mechanisms responsible for IAV pathogenesis have not yet been fully elucidated. In this study, the basic fibroblast growth factor 2 (FGF2) level was markedly increased in H1N1 virus-infected humans and mice. FGF2, which is predominately derived from epithelial cells, recruits and activates neutrophils via the FGFR2-PI3K-AKT-NFκB signaling pathway. FGF2 depletion or knockout exacerbated influenza-associated disease by impairing neutrophil recruitment and activation. More importantly, administration of the recombinant FGF2 protein significantly alleviated the severity of IAV-induced lung injury and promoted the survival of IAV-infected mice. Based on the results from experiments in which neutrophils were depleted and adoptively transferred, FGF2 protected mice against IAV infection by recruiting neutrophils. Thus, FGF2 plays a critical role in preventing IAV-induced lung injury, and FGF2 is a promising potential therapeutic target during IAV infection.
Collapse
Affiliation(s)
- Keyu Wang
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Chengcai Lai
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Tieling Li
- Chinese PLA General Hospital, Beijing, China
| | - Cheng Wang
- Chinese PLA General Hospital, Beijing, China
| | - Wei Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Bing Ni
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Changqing Bai
- Beijing 307 Hospital of PLA Affiliated with the Chinese Academy of Medical Sciences, Beijing, China
| | | | - Lina Han
- Chinese PLA General Hospital, Beijing, China
| | - Hongjing Gu
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Zhongpeng Zhao
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yueqiang Duan
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiaolan Yang
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Li Xing
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Lingna Zhao
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Shanshan Zhou
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Min Xia
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Chengyu Jiang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiliang Wang
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Penghui Yang
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China.,Beijing 302 Hospital of PLA, Beijing, China
| |
Collapse
|
38
|
Hegab AE, Ozaki M, Kameyama N, Gao J, Kagawa S, Yasuda H, Soejima K, Yin Y, Guzy RD, Nakamura Y, Ornitz DM, Betsuyaku T. Effect of FGF/FGFR pathway blocking on lung adenocarcinoma and its cancer-associated fibroblasts. J Pathol 2019; 249:193-205. [PMID: 31090071 DOI: 10.1002/path.5290] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 01/17/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are known to promote tumourigenesis through various mechanisms. Fibroblast growth factor (FGF)/FGF receptor (FGFR)-dependent lung cancers have been described. We have developed a mouse model of lung adenocarcinoma that was constructed through the induction of Fgf9 overexpression in type 2 alveolar cells. The expression of Fgf9 in adult lungs resulted in the rapid development of multiple adenocarcinoma-like tumour nodules. Here, we have characterised the contribution of CAFs and the Fgf/Fgfr signalling pathway in maintaining the lung tumours initiated by Fgf9 overexpression. We found that CAF-secreted Fgf2 contributes to tumour cell growth. CAFs overexpressed Tgfb, Mmp7, Fgf9, and Fgf2; synthesised more collagen, and secreted inflammatory cell-recruiting cytokines. CAFs also enhanced the conversion of tumour-associated macrophages (TAMs) to the tumour-supportive M2 phenotype but did not influence angiogenesis. In vivo inhibition of Fgfrs during early lung tumour development resulted in significantly smaller and fewer tumour nodules, whereas inhibition in established lung tumours caused a significant reduction in tumour size and number. Fgfr inhibition also influenced tumour stromal cells, as it significantly abolished TAM recruitment and reduced tumour vascularity. However, the withdrawal of the inhibitor caused a significant recurrence/regrowth of Fgf/Fgfr-independent lung tumours. These recurrent tumours did not possess a higher proliferative or propagative potential. Our results provide evidence that fibroblasts associated with the Fgf9-induced lung adenocarcinoma provide multiple means of support to the tumour. Although the Fgfr blocker significantly suppressed the tumour and its stromal cells, it was not sufficient to completely eliminate the tumour, probably due to the emergence of alternative (resistance/maintenance) mechanism(s). This model represents an excellent tool to further study the complex interactions between CAFs, their related chemokines, and the progression of lung adenocarcinoma; it also provides further evidence to support the need for a combinatorial strategy to treat lung cancer. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Ahmed E Hegab
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Mari Ozaki
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Naofumi Kameyama
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Jingtao Gao
- Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, PR China
| | - Shizuko Kagawa
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Yasuda
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kenzo Soejima
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yongjun Yin
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Robert D Guzy
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Yoshikazu Nakamura
- RIBOMIC Inc., Tokyo, Japan.,Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Tomoko Betsuyaku
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
39
|
Feng Y, Xu Q, Yang Y, Shi W, Meng W, Zhang H, He X, Sun M, Chen Y, Zhao J, Guo Z, Xiao K. The therapeutic effects of bone marrow-derived mesenchymal stromal cells in the acute lung injury induced by sulfur mustard. Stem Cell Res Ther 2019; 10:90. [PMID: 30867053 PMCID: PMC6416968 DOI: 10.1186/s13287-019-1189-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 01/11/2019] [Accepted: 02/25/2019] [Indexed: 12/12/2022] Open
Abstract
Background Sulfur mustard (SM) is a notorious chemical warfare agent that can cause severe acute lung injury (ALI), in addition to other lesions. Currently, effective medical countermeasures for SM are lacking. Bone marrow-derived mesenchymal stromal cells (BMSCs) possess self-renewal and multipotent differentiation capacity. BMSCs can also migrate to inflammation and injury sites and exert anti-inflammatory and tissue repair functions. Here, we report the curative effect of BMSCs on SM-induced ALI in a mouse model. Methods Mice BMSCs were injected into mice via the tail vein 24 h after SM exposure. The distribution of BMSCs in mice was detected by fluorescence imaging. The therapeutic potential of BMSCs was evaluated by the calculating survival rate. The effects of BMSCs on lung tissue injury and repair assessment were examined by staining with H&E and measuring the lung wet/dry weight ratio, BALF protein level, and respiratory function. The effects of BMSCs on the infiltration and phenotypic alteration of inflammatory cells were analyzed by immunohistochemistry and flow cytometry. The levels of chemokines and inflammatory cytokines were examined using the Luminex Performance Assay and ELISA. RNA interference, western blotting, and ELISA were applied to explore the role of the TLR4 signaling pathway in the anti-inflammatory effects of BMSCs. The extent of tissue repair was analyzed by ELISA, western blotting, and immunohistochemistry. Results Fluorescence imaging indicated that the lung is the major target organ of BMSCs after injection. The injection of BMSCs significantly improved the survival rate (p < 0.05), respiratory function, and related lung damage indexes (wet/dry weight ratio, total proteins in BALF, etc.) in mice. BMSC administration also reduced the level of pro-inflammatory cytokines, chemokines, and inflammatory cell infiltration, as well as affected the balances of M1/M2 and Th17/Treg. Furthermore, solid evidence regarding the effects of BMSCs on the increased secretion of various growth factors, the differentiation of alveolar epithelial cells, and the enhancement of cell barrier functions was also observed. Conclusion BMSCs displayed protective effects against SM-induced ALI by alleviating inflammation and promoting tissue repair. The present study provides a strong experimental basis in a mouse model and suggests possible application for future cell therapy. Electronic supplementary material The online version of this article (10.1186/s13287-019-1189-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yongwei Feng
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, China
| | - Qingqiang Xu
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, China
| | - Yuyan Yang
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, China
| | - Wenwen Shi
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, China
| | - Wenqi Meng
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, China
| | - Hao Zhang
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, China
| | - Xiaowen He
- Origincell Technology Group Co., Ltd., 1118 Halei Rd, Shanghai, 201203, China
| | - Mingxue Sun
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, China
| | - Yongchun Chen
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, China
| | - Jie Zhao
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, China
| | - Zhenhong Guo
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China.
| | - Kai Xiao
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
40
|
Blockade of platelet-derived growth factor receptor-β, not receptor-α ameliorates bleomycin-induced pulmonary fibrosis in mice. PLoS One 2018; 13:e0209786. [PMID: 30596712 PMCID: PMC6312310 DOI: 10.1371/journal.pone.0209786] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/11/2018] [Indexed: 11/19/2022] Open
Abstract
Platelet-derived growth factor (PDGF) has been implicated in the pathogenesis of pulmonary fibrosis. Nintedanib, a multi-kinase inhibitor that targets several tyrosine kinases, including PDGF receptor (PDGFR), was recently approved as an anti-fibrotic agent to reduce the deterioration of FVC in patients with idiopathic pulmonary fibrosis (IPF). However, the effects of PDGFR-α or -β on pulmonary fibrosis remain unclear. In an attempt to clarify their effects, we herein used blocking antibodies specific for PDGFR-α (APA5) and -β (APB5) in a bleomycin (BLM)-induced pulmonary fibrosis mouse model. The effects of these treatments on the growth of lung fibroblasts were examined using the 3H-thymidine incorporation assay in vitro. The anti-fibrotic effects of these antibodies were investigated with the Ashcroft score and collagen content of lungs treated with BLM. Their effects on inflammatory cells in the lungs were also analyzed using bronchoalveolar lavage fluid. We investigated damage to epithelial cells and the proliferation of fibroblasts in the lungs. APA5 and APB5 inhibited the phosphorylation of PDGFR-α and -β as well as the proliferation of lung fibroblasts induced by PDGF-AA and BB. The administration of APB5, but not APA5 effectively inhibited BLM-induced pulmonary fibrosis in mice. Apoptosis and the proliferation of epithelial cells and fibroblasts were significantly decreased by the treatment with APB5, but not by APA5. The late treatment with APB5 also ameliorated fibrosis in lungs treated with BLM. These results suggest that PDGFR-α and -β exert different effects on BLM-induced pulmonary fibrosis in mice. A specific approach using the blocking antibody for PDGFR-β may be useful for the treatment of pulmonary fibrosis.
Collapse
|
41
|
Shiraishi K, Shichino S, Ueha S, Nakajima T, Hashimoto S, Yamazaki S, Matsushima K. Mesenchymal-Epithelial Interactome Analysis Reveals Essential Factors Required for Fibroblast-Free Alveolosphere Formation. iScience 2018; 11:318-333. [PMID: 30639966 PMCID: PMC6329323 DOI: 10.1016/j.isci.2018.12.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/30/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022] Open
Abstract
Lung epithelial cells and fibroblasts are key cell populations in lung development. Fibroblasts support type 2 alveolar epithelial cells (AEC2) in the developing and mature lung. However, fibroblast-AEC2 interactions have not been clearly described. We addressed this in the present study by time course serial analysis of gene expression sequencing (SAGE-seq) of epithelial cells and fibroblasts of developing and mature murine lungs. We identified lung fibroblast-epithelial interactions that potentially regulate alveologenesis and are mediated by fibroblast-expressed ligands and epithelial cell surface receptors. In the epithelial-fibroblast co-culture alveolosphere formation assay, single intervention against fibroblast-expressed ligand or associated signaling cascades promoted or inhibited alveolosphere growth. Adding the ligand-associated molecules fibroblast growth factor 7 and Notch ligand and inhibitors of bone morphogenetic protein 4, transforming growth factor β, and glycogen synthase kinase-3β to the culture medium enabled fibroblast-free alveolosphere formation. The results revealed the essential factors regulating fibroblast-AEC2 interactions.
Collapse
Affiliation(s)
- Kazushige Shiraishi
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Noda 278-0022, Japan
| | - Shigeyuki Shichino
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Noda 278-0022, Japan
| | - Satoshi Ueha
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Noda 278-0022, Japan
| | - Takuya Nakajima
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Noda 278-0022, Japan
| | - Shinichi Hashimoto
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Noda 278-0022, Japan; Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
| | - Satoshi Yamazaki
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Kouji Matsushima
- Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Noda 278-0022, Japan.
| |
Collapse
|
42
|
Kurosawa T, Miyoshi S, Yamazaki S, Nishina T, Mikami T, Oikawa A, Homma S, Nakano H. A murine model of acute lung injury identifies growth factors to promote tissue repair and their biomarkers. Genes Cells 2018; 24:112-125. [PMID: 30474194 DOI: 10.1111/gtc.12659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 11/17/2018] [Accepted: 11/18/2018] [Indexed: 11/30/2022]
Abstract
Type II alveolar epithelial cells (AEC2s) play a crucial role in the regeneration of type I AECs after acute lung injury. The mechanisms underlying the regeneration of AEC2s are not fully understood. To address this issue, here, we investigated a murine model of acute lung injury using mice expressing human Diphtheria Toxin Receptor (DTR) under the control of Lysozyme M promoter (LysM-DTR). DT injection induced the depletion of AEC2s, alveolar macrophages, and bone marrow (BM)-derived myeloid cells in LysM-DTR mice, and the mice died within 6 days after DT injection. Apoptotic AEC2s and bronchiolar epithelial cells appeared at 24 hr, whereas Ki67-positive proliferating cells appeared in the alveoli and bronchioles in the lung of LysM-DTR mice at 72-96 hr after DT injection. Transfer of wild-type BM cells into LysM-DTR mice accelerated the regeneration of AEC2s along with the up-regulation of several growth factors. Moreover, several metabolites were significantly decreased in the sera of LysM-DTR mice compared with WT mice after DT injection, suggesting that these metabolites might be biomarkers to predict AEC2s injury. Together, LysM-DTR mice might be useful to identify growth factors to promote lung repair and the metabolites to predict the severity of lung injury.
Collapse
Affiliation(s)
- Takeyuki Kurosawa
- Department of Biochemistry, Toho University Graduate School of Medicine, Tokyo, Japan.,Department of Respiratory Medicine, Toho University Graduate School of Medicine, Tokyo, Japan.,Department of Respiratory Medicine, Omori Medical Center, Tokyo, Japan
| | - Shion Miyoshi
- Department of Biochemistry, Toho University Graduate School of Medicine, Tokyo, Japan.,Department of Respiratory Medicine, Toho University Graduate School of Medicine, Tokyo, Japan.,Department of Respiratory Medicine, Omori Medical Center, Tokyo, Japan
| | - Soh Yamazaki
- Department of Biochemistry, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Takashi Nishina
- Department of Biochemistry, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Tetuo Mikami
- Department of Pathology, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Akira Oikawa
- RIKEN Center for Sustainable Resource Science, Yokohama, Japan.,Faculty of Agriculture, Yamagata University, Tsuruoka, Japan
| | - Sakae Homma
- Department of Respiratory Medicine, Toho University Graduate School of Medicine, Tokyo, Japan.,Department of Respiratory Medicine, Omori Medical Center, Tokyo, Japan
| | - Hiroyasu Nakano
- Department of Biochemistry, Toho University Graduate School of Medicine, Tokyo, Japan.,Host Defense Research Center, Toho University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
43
|
Gonzalez Rodriguez A, Schroeder ME, Walker CJ, Anseth KS. FGF-2 inhibits contractile properties of valvular interstitial cell myofibroblasts encapsulated in 3D MMP-degradable hydrogels. APL Bioeng 2018; 2:046104. [PMID: 31069326 PMCID: PMC6481727 DOI: 10.1063/1.5042430] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/08/2018] [Indexed: 02/06/2023] Open
Abstract
Valvular interstitial cells (VICs) are responsible for the maintenance of the extracellular matrix in heart valve leaflets and, in response to injury, activate from a quiescent fibroblast to a wound healing myofibroblast phenotype. Under normal conditions, myofibroblast activation is transient, but the chronic presence of activated VICs can lead to valve diseases, such as fibrotic aortic valve stenosis, for which non-surgical treatments remain elusive. We monitored the porcine VIC response to exogenously delivered fibroblast growth factor 2 (FGF-2; 100 ng/ml), transforming growth factor beta 1 (TGF-β1; 5 ng/ml), or a combination of the two while cultured within 3D matrix metalloproteinase (MMP)-degradable 8-arm 40 kDa poly(ethylene glycol) hydrogels that mimic aspects of the aortic valve. Here, we aimed to investigate VIC myofibroblast activation and subsequent contraction or the reparative wound healing response. To this end, VIC morphology, proliferation, gene expression related to the myofibroblast phenotype [alpha smooth muscle actin (α-SMA) and connective tissue growth factor (CTGF)] and matrix remodeling [collagens (COL1A1 and COL3) and MMP1], and contraction assays were used to quantify the cell response. Treatment with FGF-2 resulted in increased cellular proliferation while reducing the myofibroblast phenotype, as seen by decreased expression of CTGF and α-SMA, and reduced contraction relative to untreated control, suggesting that FGF-2 encourages a reparative phenotype, even in the presence of TGF-β1. TGF-β1 treatment predictably led to an increased proportion of VICs exhibiting the myofibroblast phenotype, indicated by the presence of α-SMA, increased gene expression indicative of matrix remodeling, and bulk contraction of the hydrogels. Functional contraction assays and biomechanical analyses were performed on VIC encapsulated hydrogels and porcine aortic valve tissue explants to validate these findings.
Collapse
|
44
|
Koo HY, El-Baz LM, House SL, Cilvik SN, Dorry SJ, Shoukry NM, Salem ML, Hafez HS, Dulin NO, Ornitz DM, Guzy RD. Fibroblast growth factor 2 decreases bleomycin-induced pulmonary fibrosis and inhibits fibroblast collagen production and myofibroblast differentiation. J Pathol 2018; 246:54-66. [PMID: 29873400 PMCID: PMC6175645 DOI: 10.1002/path.5106] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 04/12/2018] [Accepted: 05/19/2018] [Indexed: 02/05/2023]
Abstract
Fibroblast growth factor (FGF) signaling has been implicated in the pathogenesis of pulmonary fibrosis. Mice lacking FGF2 have increased mortality and impaired epithelial recovery after bleomycin exposure, supporting a protective or reparative function following lung injury. To determine whether FGF2 overexpression reduces bleomycin-induced injury, we developed an inducible genetic system to express FGF2 in type II pneumocytes. Double-transgenic (DTG) mice with doxycycline-inducible overexpression of human FGF2 (SPC-rtTA;TRE-hFGF2) or single-transgenic controls were administered intratracheal bleomycin and fed doxycycline chow, starting at either day 0 or day 7. In addition, wild-type mice received intratracheal or intravenous recombinant FGF2, starting at the time of bleomycin treatment. Compared to controls, doxycycline-induced DTG mice had decreased pulmonary fibrosis 21 days after bleomycin, as assessed by gene expression and histology. This beneficial effect was seen when FGF2 overexpression was induced at day 0 or day 7 after bleomycin. FGF2 overexpression did not alter epithelial gene expression, bronchoalveolar lavage cellularity or total protein. In vitro studies using primary mouse and human lung fibroblasts showed that FGF2 strongly inhibited baseline and TGFβ1-induced expression of alpha smooth muscle actin (αSMA), collagen, and connective tissue growth factor. While FGF2 did not suppress phosphorylation of Smad2 or Smad-dependent gene expression, FGF2 inhibited TGFβ1-induced stress fiber formation and serum response factor-dependent gene expression. FGF2 inhibition of stress fiber formation and αSMA requires FGF receptor 1 (FGFR1) and downstream MEK/ERK, but not AKT signaling. In summary, overexpression of FGF2 protects against bleomycin-induced pulmonary fibrosis in vivo and reverses TGFβ1-induced collagen and αSMA expression and stress fiber formation in lung fibroblasts in vitro, without affecting either inflammation or epithelial gene expression. Our results suggest that in the lung, FGF2 is antifibrotic in part through decreased collagen expression and fibroblast to myofibroblast differentiation. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Hyun Young Koo
- University of Chicago, Department of Medicine, Section of Pulmonary and Critical Care Medicine, Chicago, IL, USA
| | - Lamis M.F. El-Baz
- University of Chicago, Department of Medicine, Section of Pulmonary and Critical Care Medicine, Chicago, IL, USA
- Suez University, Faculty of Science, Zoology Department, Suez, Egypt
| | - Stacey L. House
- Washington University School of Medicine, Department of Emergency Medicine, St. Louis, MO, USA
| | - Sarah N. Cilvik
- Washington University School of Medicine, Department of Developmental Biology, St. Louis, MO, USA
| | - Samuel J. Dorry
- University of Chicago, Department of Medicine, Section of Pulmonary and Critical Care Medicine, Chicago, IL, USA
| | - Nahla M. Shoukry
- Suez University, Faculty of Science, Zoology Department, Suez, Egypt
| | - Mohamed L. Salem
- Tanta University, Center of Excellence in Cancer Research, Faculty of Science, Immunology & Biotechnology Department, Tanta, Egypt
| | - Hani S. Hafez
- Suez University, Faculty of Science, Zoology Department, Suez, Egypt
| | - Nickolai O. Dulin
- University of Chicago, Department of Medicine, Section of Pulmonary and Critical Care Medicine, Chicago, IL, USA
| | - David M. Ornitz
- Washington University School of Medicine, Department of Developmental Biology, St. Louis, MO, USA
| | - Robert D. Guzy
- University of Chicago, Department of Medicine, Section of Pulmonary and Critical Care Medicine, Chicago, IL, USA
| |
Collapse
|
45
|
Chanda D, Otoupalova E, Smith SR, Volckaert T, De Langhe SP, Thannickal VJ. Developmental pathways in the pathogenesis of lung fibrosis. Mol Aspects Med 2018; 65:56-69. [PMID: 30130563 DOI: 10.1016/j.mam.2018.08.004] [Citation(s) in RCA: 327] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/17/2018] [Indexed: 12/20/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and terminal lung disease with no known cure. IPF is a disease of aging, with median age of diagnosis over 65 years. Median survival is between 3 and 5 years after diagnosis. IPF is characterized primarily by excessive deposition of extracellular matrix (ECM) proteins by activated lung fibroblasts and myofibroblasts, resulting in reduced gas exchange and impaired pulmonary function. Growing evidence supports the concept of a pro-fibrotic environment orchestrated by underlying factors such as genetic predisposition, chronic injury and aging, oxidative stress, and impaired regenerative responses may account for disease development and persistence. Currently, two FDA approved drugs have limited efficacy in the treatment of IPF. Many of the genes and gene networks associated with lung development are induced or activated in IPF. In this review, we analyze current knowledge in the field, gained from both basic and clinical research, to provide new insights into the disease process, and potential approaches to treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Diptiman Chanda
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Eva Otoupalova
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Samuel R Smith
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Thomas Volckaert
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Stijn P De Langhe
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
46
|
Hosseinzadeh A, Javad-Moosavi SA, Reiter RJ, Hemati K, Ghaznavi H, Mehrzadi S. Idiopathic pulmonary fibrosis (IPF) signaling pathways and protective roles of melatonin. Life Sci 2018; 201:17-29. [PMID: 29567077 DOI: 10.1016/j.lfs.2018.03.032] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/13/2018] [Accepted: 03/15/2018] [Indexed: 12/19/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by the progressive loss of lung function due to tissue scarring. A variety of pro-inflammatory and pro-fibrogenic factors including interleukin‑17A, transforming growth factor β, Wnt/β‑catenin, vascular endothelial growth factor, platelet-derived growth factor, fibroblast growth factors, endotelin‑1, renin angiotensin system and impaired caveolin‑1 function are involved in the IPF pathogenesis. Current therapies for IPF have some limitations and this highlights the need for effective therapeutic agents to treat this fatal disease. Melatonin and its metabolites are broad-spectrum antioxidants that not only remove reactive oxygen and nitrogen species by radical scavenging but also up-regulate the expression and activity of endogenous antioxidants. Via these actions, melatonin and its metabolites modulate a variety of molecular pathways in different pathophysiological conditions. Herein, we review the signaling pathways involved in the pathophysiology of IPF and the potentially protective effects of melatonin on these pathways.
Collapse
Affiliation(s)
- Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health, San Antonio, TX, USA
| | - Karim Hemati
- Department of Anesthesiology, Iran University of Medical Sciences, Tehran, Iran; Department of Anesthesiology, Ilam University of Medical Sciences, Ilam, Iran
| | - Habib Ghaznavi
- Department of Pharmacology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
47
|
Ialenti A, Caiazzo E, Morello S, Carnuccio R, Cicala C. Adenosine A 2A Receptor Agonist, 2- p-(2-Carboxyethyl)phenethylamino-5'- N-ethylcarboxamidoadenosine Hydrochloride Hydrate, Inhibits Inflammation and Increases Fibroblast Growth Factor-2 Tissue Expression in Carrageenan-Induced Rat Paw Edema. J Pharmacol Exp Ther 2018; 364:221-228. [PMID: 29212832 DOI: 10.1124/jpet.117.244319] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/27/2017] [Indexed: 03/08/2025] Open
Abstract
Adenosine is the final product of ATP metabolism, mainly derived from the action of 5'-nucleotidase cleavage of AMP. Cellular production of adenosine is greatly enhanced in inflamed tissues, ischemic tissues, and under hypoxia, where ATP is released from damaged cells. Much evidence has been accumulated on adenosine anti-inflammatory effects mediated through A2A receptor activation; A2A adenosine receptor has also been shown to play a role in matrix deposition and wound healing in a damaged tissue, contributing to dermal tissue protection and repair. Fibroblast growth factor-2 (FGF-2) is a powerful mitogen for fibroblast; it is expressed by several inflammatory cell types and plays a pivotal role in angiogenesis, wound healing, gastric ulcer protection. Human recombinant FGF-2 has been shown to have anti-inflammatory effects. The purpose of the present work was to investigate on the anti-inflammatory effect of systemic administration of the adenosine A2A agonist 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamidoadenosine hydrochloride hydrate (CGS21680) in the rat model of carrageenan-induced paw edema. We found that CGS21680 inhibits inflammation induced by carrageenan injection into the rat paw, and this effect is associated to the local reduction of cytokine levels and dermal increase of FGF-2 expression. Our results suggest that FGF-2 might be involved in the anti-inflammatory and tissue protective effect due to A2A receptor activation.
Collapse
Affiliation(s)
- Armando Ialenti
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy (A.I., E.C., R.C., C.C.) and Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy (S.M.)
| | - Elisabetta Caiazzo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy (A.I., E.C., R.C., C.C.) and Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy (S.M.)
| | - Silvana Morello
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy (A.I., E.C., R.C., C.C.) and Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy (S.M.)
| | - Rosa Carnuccio
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy (A.I., E.C., R.C., C.C.) and Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy (S.M.)
| | - Carla Cicala
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy (A.I., E.C., R.C., C.C.) and Department of Pharmacy, University of Salerno, Fisciano, Salerno, Italy (S.M.)
| |
Collapse
|
48
|
Li X, Li X, Zhang Q, Zhao T. Low molecular weight fucoidan and its fractions inhibit renal epithelial mesenchymal transition induced by TGF-β1 or FGF-2. Int J Biol Macromol 2017. [DOI: 10.1016/j.ijbiomac.2017.06.058] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
49
|
Maddaluno L, Urwyler C, Werner S. Fibroblast growth factors: key players in regeneration and tissue repair. Development 2017; 144:4047-4060. [PMID: 29138288 DOI: 10.1242/dev.152587] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tissue injury initiates a complex repair process, which in some organisms can lead to the complete regeneration of a tissue. In mammals, however, the repair of most organs is imperfect and results in scar formation. Both regeneration and repair are orchestrated by a highly coordinated interplay of different growth factors and cytokines. Among the key players are the fibroblast growth factors (FGFs), which control the migration, proliferation, differentiation and survival of different cell types. In addition, FGFs influence the expression of other factors involved in the regenerative response. Here, we summarize current knowledge on the roles of endogenous FGFs in regeneration and repair in different organisms and in different tissues and organs. Gaining a better understanding of these FGF activities is important for appropriate modulation of FGF signaling after injury to prevent impaired healing and to promote organ regeneration in humans.
Collapse
Affiliation(s)
- Luigi Maddaluno
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Corinne Urwyler
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| |
Collapse
|
50
|
Worke LJ, Barthold JE, Seelbinder B, Novak T, Main RP, Harbin SL, Neu CP. Densification of Type I Collagen Matrices as a Model for Cardiac Fibrosis. Adv Healthc Mater 2017; 6. [PMID: 28881428 DOI: 10.1002/adhm.201700114] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/10/2017] [Indexed: 12/17/2022]
Abstract
Cardiac fibrosis is a disease state characterized by excessive collagenous matrix accumulation within the myocardium that can lead to ventricular dilation and systolic failure. Current treatment options are severely lacking due in part to the poor understanding of the complexity of molecular pathways involved in cardiac fibrosis. To close this gap, in vitro model systems that recapitulate the defining features of the fibrotic cellular environment are in need. Type I collagen, a major cardiac extracellular matrix protein and the defining component of fibrotic depositions, is an attractive choice for a fibrosis model, but demonstrates poor mechanical strength due to solubility limits. However, plastic compression of collagen matrices is shown to significantly increase its mechanical properties. Here, confined compression of oligomeric, type I collagen matrices is utilized to resemble defining hallmarks seen in fibrotic tissue such as increased collagen content, fibril thickness, and bulk compressive modulus. Cardiomyocytes seeded on compressed matrices show a strong beating abrogation as observed in cardiac fibrosis. Gene expression analysis of selected fibrosis markers indicates fibrotic activation and cardiomyocyte maturation with regard to the existing literature. With these results, a promising first step toward a facile heart-on-chip model is presented to study cardiac fibrosis.
Collapse
Affiliation(s)
- Logan J. Worke
- Weldon School of Biomedical Engineering; Purdue University; West Lafayette IN USA 47906
| | - Jeanne E. Barthold
- Department of Mechanical Engineering; University of Colorado Boulder; Boulder CO USA 80309
| | - Benjamin Seelbinder
- Department of Mechanical Engineering; University of Colorado Boulder; Boulder CO USA 80309
| | - Tyler Novak
- Weldon School of Biomedical Engineering; Purdue University; West Lafayette IN USA 47906
| | - Russell P. Main
- Weldon School of Biomedical Engineering; Purdue University; West Lafayette IN USA 47906
- Department of Basic Medical Sciences; Purdue University; West Lafayette IN USA 47906
| | - Sherry L. Harbin
- Weldon School of Biomedical Engineering; Purdue University; West Lafayette IN USA 47906
- Department of Basic Medical Sciences; Purdue University; West Lafayette IN USA 47906
| | - Corey P. Neu
- Weldon School of Biomedical Engineering; Purdue University; West Lafayette IN USA 47906
- Department of Mechanical Engineering; University of Colorado Boulder; Boulder CO USA 80309
| |
Collapse
|