101
|
Li YL, Chang JT, Lee LY, Fan KH, Lu YC, Li YC, Chiang CH, You GR, Chen HY, Cheng AJ. GDF15 contributes to radioresistance and cancer stemness of head and neck cancer by regulating cellular reactive oxygen species via a SMAD-associated signaling pathway. Oncotarget 2017; 8:1508-1528. [PMID: 27903972 PMCID: PMC5352073 DOI: 10.18632/oncotarget.13649] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 11/12/2016] [Indexed: 12/22/2022] Open
Abstract
Radiotherapy is an integral part for the treatment of head and neck cancer (HNC), while radioresistance is a major cause leads to treatment failure. GDF15, a member of the TGF-β superfamily, is hypothesized to participate in various types of homeostasis. However, the potential role of this molecule in regulation of radiosensitivity remains unclear. In this study, we demonstrated that GDF15 contributed to radioresistance of HNC, as determined by both gain- and lost-of-functional experiments. These results were achieved by the induction of mitochondrial membrane potential and suppression of intracellular reactive oxygen species (ROS). We further showed that GDF15 facilitated the conversion of cancer stemness, as assessed by the promotion of CD44+ and ALDH1+ cell populations and spheroid cell formation. At molecular level, GDF15 conferred to these cellular functions was through phosphorylated SMAD1 proteins to elite downstream signaling molecules. These cellular results were further confirmed in a tumor xenograft mouse study. Taken together, our results demonstrated that GDF15 contributed to radioresistance and cancer stemness by regulating cellular ROS levels via a SMAD-associated signaling pathway. GDF15 may serve as a prediction marker of radioresistance and a therapeutic target for the development of radio-sensitizing agents for the treatment of refractory HNC.
Collapse
Affiliation(s)
- Yan-Liang Li
- Department of Medical Biotechnology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Joseph T. Chang
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Kang-Hsing Fan
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Ya-Ching Lu
- Department of Medical Biotechnology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Yi-Chen Li
- Department of Medical Biotechnology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chang-Hsu Chiang
- Department of Medical Biotechnology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Guo-Rung You
- Department of Medical Biotechnology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Hsin-Ying Chen
- Department of Medical Biotechnology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Ann-Joy Cheng
- Department of Medical Biotechnology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| |
Collapse
|
102
|
John AE, Porte J, Jenkins G, Tatler AL. Methods for the Assessment of Active Transforming Growth Factor-β in Cells and Tissues. Methods Mol Biol 2017; 1627:351-365. [PMID: 28836213 DOI: 10.1007/978-1-4939-7113-8_23] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The potent and pluripotent cytokine TGFβ has important roles in normal homeostasis and disease pathogenesis. Once released from cells, TGFβ exists in both latent and functionally active forms. Large amounts of latent TGFβ are secreted from cells and sequestered in extracellular matrix, only a small proportion of which is activated at any given time. Accurate assessment of TGFβ activity levels is an important measurement in biological research and requires methods distinct from measuring total levels of TGFβ expression as small changes in TGFβ activity levels could be masked by the large amounts of latent TGFβ available to be measured. In this chapter, we describe detailed experimental methods for assessing levels of active TGFβ in cells and tissues. This chapter includes methods for the assessment of TGFβ activity in cells in vitro, in ex vivo precision cut tissue, and in vivo.
Collapse
Affiliation(s)
- Alison E John
- Division of Respiratory Medicine, School of Medicine, Nottingham City Hospital, University of Nottingham, Nottingham, UK
| | - Joanne Porte
- Division of Respiratory Medicine, School of Medicine, Nottingham City Hospital, University of Nottingham, Nottingham, UK
| | - Gisli Jenkins
- Division of Respiratory Medicine, School of Medicine, Nottingham City Hospital, University of Nottingham, Nottingham, UK
| | - Amanda L Tatler
- Division of Respiratory Medicine, School of Medicine, Nottingham City Hospital, University of Nottingham, Nottingham, UK.
| |
Collapse
|
103
|
Burgy O, Wettstein G, Bellaye PS, Decologne N, Racoeur C, Goirand F, Beltramo G, Hernandez JF, Kenani A, Camus P, Bettaieb A, Garrido C, Bonniaud P. Deglycosylated bleomycin has the antitumor activity of bleomycin without pulmonary toxicity. Sci Transl Med 2016; 8:326ra20. [PMID: 26888428 DOI: 10.1126/scitranslmed.aad7785] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Bleomycin (BLM) is a potent anticancer drug used to treat different malignancies, mainly lymphomas, germ cell tumors, and melanomas. Unfortunately, BLM has major, dose-dependent, pulmonary toxicity that affects 20% of treated individuals. The most severe form of BLM-induced pulmonary toxicity is lung fibrosis. Deglyco-BLM is a molecule derived from BLM in which the sugar residue d-mannosyl-l-glucose disaccharide has been deleted. The objective of this study was to assess the anticancer activity and lung toxicity of deglyco-BLM. We compared the antitumor activity and pulmonary toxicity of intraperitoneally administrated deglyco-BLM and BLM in three rodent models. Pulmonary toxicity was examined in depth after intratracheal administration of both chemotherapeutic agents. The effect of both drugs was further studied in epithelial alveolar cells in vitro. We demonstrated in rodent cancer models, including a human Hodgkin's lymphoma xenograft and a syngeneic melanoma model, that intraperitoneal deglyco-BLM is as effective as BLM in inducing tumor regression. Whereas the antitumor effect of BLM was accompanied by a loss of body weight and the development of pulmonary toxicity, deglyco-BLM did not affect body weight and did not engender lung injury. Both molecules induced lung epithelial cell apoptosis after intratracheal administration, but deglyco-BLM lost the ability to induce caspase-1 activation and the production of ROS (reactive oxygen species), transforming growth factor-β1, and other profibrotic and inflammatory cytokines in the lungs of mice and in vitro. Deglyco-BLM should be considered for clinical testing as a less toxic alternative to BLM in cancer therapy.
Collapse
Affiliation(s)
- Olivier Burgy
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France
| | - Guillaume Wettstein
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France
| | - Pierre S Bellaye
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France
| | - Nathalie Decologne
- Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France. EPHE, Tumor Immunology and Immunotherapy Laboratory, Dijon 21079, France
| | - Cindy Racoeur
- Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France. EPHE, Tumor Immunology and Immunotherapy Laboratory, Dijon 21079, France
| | - Françoise Goirand
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France
| | - Guillaume Beltramo
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France. Service de Pneumologie et Soins Intensifs Respiratoires, Centre Hospitalier Universitaire (CHU), Dijon 21079, France
| | - Jean-François Hernandez
- Institut des Biomolécules Max Mousseron, Faculty of Pharmacy, University of Montpellier, Montpellier 34093, France
| | - Abderraouf Kenani
- Department of Biochemistry, University of Monastir, Monastir 5000, Tunisia
| | - Philippe Camus
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France. Service de Pneumologie et Soins Intensifs Respiratoires, Centre Hospitalier Universitaire (CHU), Dijon 21079, France
| | - Ali Bettaieb
- Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France. EPHE, Tumor Immunology and Immunotherapy Laboratory, Dijon 21079, France
| | - Carmen Garrido
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France. Anticancer Centre Georges François Leclerc, CGFL, Dijon 21079, France
| | - Philippe Bonniaud
- INSERM, LNC UMR 866, Laboratoire d'Excellence LipSTIC, Dijon 21079, France. Equipe "Heat Shock Proteins" Labellisée par la Ligue Nationale contre le Cancer, Dijon 21079, France. Faculty of Medicine and Pharmacy of Dijon, Université Bourgogne Franche-Comté, Dijon 21079, France. Service de Pneumologie et Soins Intensifs Respiratoires, Centre Hospitalier Universitaire (CHU), Dijon 21079, France.
| |
Collapse
|
104
|
Tsui P, Higazi DR, Wu Y, Dunmore R, Solier E, Kasali T, Bond NJ, Huntington C, Carruthers A, Hood J, Borrok MJ, Barnes A, Rickert K, Phipps S, Shirinian L, Zhu J, Bowen MA, Dall'Acqua W, Murray LA. The TGF-β inhibitory activity of antibody 37E1B5 depends on its H-CDR2 glycan. MAbs 2016; 9:104-113. [PMID: 27834568 DOI: 10.1080/19420862.2016.1255390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Excessive transforming growth factor (TGF)-β is associated with pro-fibrotic responses in lung disease, yet it also plays essential roles in tissue homeostasis and autoimmunity. Therefore, selective inhibition of excessive and aberrant integrin-mediated TGF-β activation via targeting the α-v family of integrins is being pursued as a therapeutic strategy for chronic lung diseases, to mitigate any potential safety concerns with global TGF-β inhibition. In this work, we reveal a novel mechanism of inhibiting TGF-β activation utilized by an αvβ8 targeting antibody, 37E1B5. This antibody blocks TGF-β activation while not inhibiting cell adhesion. We show that an N-linked complex-type Fab glycan in H-CDR2 of 37E1B5 is directly involved in the inhibition of latent TGF-β activation. Removal of the Fab N-glycosylation site by single amino acid substitution, or removal of N-linked glycans by enzymatic digestion, drastically reduced the antibody's ability to inhibit latency-associated peptide (LAP) and αvβ8 association, and TGF-β activation in an αvβ8-mediated TGF-β signaling reporter assay. Our results indicate a non-competitive, allosteric inhibition of 37E1B5 on αvβ8-mediated TGF-β activation. This unique, H-CDR2 glycan-mediated mechanism may account for the potent but tolerable TGF-b activation inhibition and lack of an effect on cellular adhesion by the antibody.
Collapse
Affiliation(s)
- Ping Tsui
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Daniel R Higazi
- b Biopharmaceutical Development, MedImmune Ltd , Cambridge , UK
| | - Yanli Wu
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Rebecca Dunmore
- c Respiratory, Inflammation and Autoimmunity, MedImmune Ltd , Cambridge , UK
| | - Emilie Solier
- b Biopharmaceutical Development, MedImmune Ltd , Cambridge , UK
| | - Toyin Kasali
- b Biopharmaceutical Development, MedImmune Ltd , Cambridge , UK
| | - Nicholas J Bond
- b Biopharmaceutical Development, MedImmune Ltd , Cambridge , UK
| | | | - Alan Carruthers
- c Respiratory, Inflammation and Autoimmunity, MedImmune Ltd , Cambridge , UK
| | - John Hood
- e Translational Sciences, Medimmune Ltd ., Cambridge , UK
| | - M Jack Borrok
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Arnita Barnes
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Keith Rickert
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Sandrina Phipps
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Lena Shirinian
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Jie Zhu
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Michael A Bowen
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - William Dall'Acqua
- a Antibody Discovery and Protein Engineering, Medimmune LLC , Gaithersburg , MD , USA
| | - Lynne A Murray
- c Respiratory, Inflammation and Autoimmunity, MedImmune Ltd , Cambridge , UK
| |
Collapse
|
105
|
Sulaiman AA, Zolnierczyk K, Japa O, Owen JP, Maddison BC, Emes RD, Hodgkinson JE, Gough KC, Flynn RJ. A Trematode Parasite Derived Growth Factor Binds and Exerts Influences on Host Immune Functions via Host Cytokine Receptor Complexes. PLoS Pathog 2016; 12:e1005991. [PMID: 27806135 PMCID: PMC5091765 DOI: 10.1371/journal.ppat.1005991] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 10/11/2016] [Indexed: 12/03/2022] Open
Abstract
The trematode Fasciola hepatica is responsible for chronic zoonotic infection globally. Despite causing a potent T-helper 2 response, it is believed that potent immunomodulation is responsible for rendering this host reactive non-protective host response thereby allowing the parasite to remain long-lived. We have previously identified a growth factor, FhTLM, belonging to the TGF superfamily can have developmental effects on the parasite. Herein we demonstrate that FhTLM can exert influence over host immune functions in a host receptor specific fashion. FhTLM can bind to receptor members of the Transforming Growth Factor (TGF) superfamily, with a greater affinity for TGF-β RII. Upon ligation FhTLM initiates the Smad2/3 pathway resulting in phenotypic changes in both fibroblasts and macrophages. The formation of fibroblast CFUs is reduced when cells are cultured with FhTLM, as a result of TGF-β RI kinase activity. In parallel the wound closure response of fibroblasts is also delayed in the presence of FhTLM. When stimulated with FhTLM blood monocyte derived macrophages adopt an alternative or regulatory phenotype. They express high levels interleukin (IL)-10 and arginase-1 while displaying low levels of IL-12 and nitric oxide. Moreover they also undergo significant upregulation of the inhibitory receptor PD-L1 and the mannose receptor. Use of RNAi demonstrates that this effect is dependent on TGF-β RII and mRNA knock-down leads to a loss of IL-10 and PD-L1. Finally, we demonstrate that FhTLM aids newly excysted juveniles (NEJs) in their evasion of antibody-dependent cell cytotoxicity (ADCC) by reducing the NO response of macrophages—again dependent on TGF-β RI kinase. FhTLM displays restricted expression to the F. hepatica gut resident NEJ stages. The altered fibroblast responses would suggest a role for dampened tissue repair responses in facilitating parasite migration. Furthermore, the adoption of a regulatory macrophage phenotype would allow for a reduced effector response targeting juvenile parasites which we demonstrate extends to an abrogation of the ADCC response. Thus suggesting that FhTLM is a stage specific evasion molecule that utilises host cytokine receptors. These findings are the first to clearly demonstrate the interaction of a helminth cytokine with a host receptor complex resulting in immune modifications that facilitate the non-protective chronic immune response which is characteristic of F. hepatica infection. Parasitic worms, helminths, can cause long-lived chronic infection in many hosts that they infection. The liver fluke, Fasciola hepatica, is one such parasite causing global infection of both humans and animals. F. hepatica exerts an influence over the immune system such that it avoids effector mechanisms and prevents the development of effective immunity. Here we characterise a molecule—FhTLM—derived from juvenile parasites that is similar to the regulatory cytokine TGF-β. We show that FhTLM will bind to host TGF-β receptors with a reduced affinity when compared with mammalian TGF-β. Despite this FhTLM can induce Smad2/3 signalling in host leukocytes, which is key to initiating gene transcription. Phenotypically FhTLM causes fibroblasts to slow their growth and replication response resulting in slower wound healing. Importantly FhTLM induces a macrophage phenotype that resembles a regulatory macrophage phenotype identified in other species undergoing helminth infection. Finally we Our work highlights the potential of FhTLM to play important roles in controlling host immunity when initially infected with juvenile parasites, thereby preventing the development of effective immunity.
Collapse
Affiliation(s)
- Azad A. Sulaiman
- School of Veterinary Medicine and Science, the University of Nottingham, Sutton Bonington Campus, Nottingham, United Kingdom
| | - Katarzyna Zolnierczyk
- School of Veterinary Medicine and Science, the University of Nottingham, Sutton Bonington Campus, Nottingham, United Kingdom
| | - Ornampai Japa
- School of Veterinary Medicine and Science, the University of Nottingham, Sutton Bonington Campus, Nottingham, United Kingdom
- School of Medicine, University of Phayao, Phayao, Thailand
| | - Jonathan P. Owen
- ADAS UK, the University of Nottingham, Sutton Bonington Campus, Nottingham, United Kingdom
| | - Ben C. Maddison
- ADAS UK, the University of Nottingham, Sutton Bonington Campus, Nottingham, United Kingdom
| | - Richard D. Emes
- School of Veterinary Medicine and Science, the University of Nottingham, Sutton Bonington Campus, Nottingham, United Kingdom
| | - Jane E. Hodgkinson
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Kevin C. Gough
- School of Veterinary Medicine and Science, the University of Nottingham, Sutton Bonington Campus, Nottingham, United Kingdom
| | - Robin J. Flynn
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| |
Collapse
|
106
|
Burhan I, Furini G, Lortat-Jacob H, Atobatele AG, Scarpellini A, Schroeder N, Atkinson J, Maamra M, Nutter FH, Watson P, Vinciguerra M, Johnson TS, Verderio EAM. Interplay between transglutaminases and heparan sulphate in progressive renal scarring. Sci Rep 2016; 6:31343. [PMID: 27694984 PMCID: PMC5046136 DOI: 10.1038/srep31343] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 07/18/2016] [Indexed: 01/19/2023] Open
Abstract
Transglutaminase-2 (TG2) is a new anti-fibrotic target for chronic kidney disease, for its role in altering the extracellular homeostatic balance leading to excessive build-up of matrix in kidney. However, there is no confirmation that TG2 is the only transglutaminase involved, neither there are strategies to control its action specifically over that of the conserved family-members. In this study, we have profiled transglutaminase isozymes in the rat subtotal nephrectomy (SNx) model of progressive renal scarring. All transglutaminases increased post-SNx peaking at loss of renal function but TG2 was the predominant enzyme. Upon SNx, extracellular TG2 deposited in the tubulointerstitium and peri-glomerulus via binding to heparan sulphate (HS) chains of proteoglycans and co-associated with syndecan-4. Extracellular TG2 was sufficient to activate transforming growth factor-β1 in tubular epithelial cells, and this process occurred in a HS-dependent way, in keeping with TG2-affinity for HS. Analysis of heparin binding of the main transglutaminases revealed that although the interaction between TG1 and HS is strong, the conformational heparin binding site of TG2 is not conserved, suggesting that TG2 has a unique interaction with HS within the family. Our data provides a rationale for a novel anti-fibrotic strategy specifically targeting the conformation-dependent TG2-epitope interacting with HS.
Collapse
Affiliation(s)
- Izhar Burhan
- Nottingham Trent University, School of Science and Technology, Nottingham, NG11 8NS, United Kingdom
| | - Giulia Furini
- Nottingham Trent University, School of Science and Technology, Nottingham, NG11 8NS, United Kingdom
| | - Hugues Lortat-Jacob
- Institut de Biologie Structurale, UMR 5075, Univ. Grenoble Alpes, CNRS, CEA, Grenoble, F-38027, France
| | - Adeola G. Atobatele
- Nottingham Trent University, School of Science and Technology, Nottingham, NG11 8NS, United Kingdom
| | - Alessandra Scarpellini
- Nottingham Trent University, School of Science and Technology, Nottingham, NG11 8NS, United Kingdom
| | - Nina Schroeder
- Nottingham Trent University, School of Science and Technology, Nottingham, NG11 8NS, United Kingdom
| | - John Atkinson
- University of Sheffield, Academic Nephrology Unit, Medical School, Sheffield, S10 2RZ, United Kingdom
| | - Mabrouka Maamra
- University of Sheffield, Academic Nephrology Unit, Medical School, Sheffield, S10 2RZ, United Kingdom
| | - Faith H. Nutter
- University of Sheffield, Academic Nephrology Unit, Medical School, Sheffield, S10 2RZ, United Kingdom
| | - Philip Watson
- University of Sheffield, Academic Nephrology Unit, Medical School, Sheffield, S10 2RZ, United Kingdom
| | - Manlio Vinciguerra
- Nottingham Trent University, School of Science and Technology, Nottingham, NG11 8NS, United Kingdom
| | - Timothy S. Johnson
- University of Sheffield, Academic Nephrology Unit, Medical School, Sheffield, S10 2RZ, United Kingdom
| | | |
Collapse
|
107
|
Ghebes CA, van Lente J, Post JN, Saris DBF, Fernandes H. High-Throughput Screening Assay Identifies Small Molecules Capable of Modulating the BMP-2 and TGF-β1 Signaling Pathway. SLAS DISCOVERY 2016; 22:40-50. [PMID: 27628690 DOI: 10.1177/1087057116669346] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Modulating the bone morphogenetic protein 2 (BMP-2) and transforming growth factor-β1 (TGF-β1) signaling pathways is essential during tendon/ligament (T/L) healing. Unfortunately, growth factor delivery in situ is far from trivial and, in many cases, the necessary growth factors are not approved for clinical use. Here we used a BMP-2 and a TGF-β1 reporter cell line to screen a library of 1280 Food and Drug Administration-approved small molecules and identify modulators of both signaling pathways. We identified four compounds capable of modulating BMP and TGF signaling on primary human tendon-derived cells (huTCs) and describe their effects on proliferation and differentiation of these cells.
Collapse
Affiliation(s)
- Corina-Adriana Ghebes
- 1 MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Jéré van Lente
- 1 MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Janine Nicole Post
- 1 MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Daniel B F Saris
- 1 MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands.,2 Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hugo Fernandes
- 1 MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands.,3 Center for Neuroscience and Cell Biology (CNC), Stem Cells and Drug Screening Lab, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
108
|
Tomasovic A, Kurrle N, Wempe F, De-Zolt S, Scheibe S, Koli K, Serchinger M, Schnütgen F, Sürün D, Sterner-Kock A, Weissmann N, von Melchner H. Ltbp4 regulates Pdgfrβ expression via TGFβ-dependent modulation of Nrf2 transcription factor function. Matrix Biol 2016; 59:109-120. [PMID: 27645114 DOI: 10.1016/j.matbio.2016.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 09/07/2016] [Accepted: 09/11/2016] [Indexed: 10/21/2022]
Abstract
Latent transforming growth factor beta binding protein 4 (LTBP4) belongs to the fibrillin/LTBP family of proteins and plays an important role as a structural component of extracellular matrix (ECM) and local regulator of TGFβ signaling. We have previously reported that Ltbp4S knock out mice (Ltbp4S-/-) develop centrilobular emphysema reminiscent of late stage COPD, which could be partially rescued by inactivating the antioxidant protein Sestrin 2 (Sesn2). More recent studies showed that Sesn2 knock out mice upregulate Pdgfrβ-controlled alveolar maintenance programs that protect against cigarette smoke induced pulmonary emphysema. Based on this, we hypothesized that the emphysema of Ltbp4S-/- mice is primarily caused by defective Pdgfrβ signaling. Here we show that LTBP4 induces Pdgfrβ signaling by inhibiting the antioxidant Nrf2/Keap1 pathway in a TGFβ-dependent manner. Overall, our data identified Ltbp4 as a major player in lung remodeling and injury repair.
Collapse
Affiliation(s)
- Ana Tomasovic
- Department of Molecular Hematology, Goethe University Medical School, D-60590 Frankfurt am Main, Germany
| | - Nina Kurrle
- Department of Molecular Hematology, Goethe University Medical School, D-60590 Frankfurt am Main, Germany
| | - Frank Wempe
- Department of Molecular Hematology, Goethe University Medical School, D-60590 Frankfurt am Main, Germany
| | - Silke De-Zolt
- Department of Molecular Hematology, Goethe University Medical School, D-60590 Frankfurt am Main, Germany
| | - Susan Scheibe
- Excellence Cluster Cardiopulmonary System (ECCPS), Justus-Liebig-University Giessen, Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), D-35392 Giessen, Germany
| | - Katri Koli
- Research Programs Unit and Transplantation Laboratory, Haartman Institute, University of Helsinki, 00014, Helsinki, Finland
| | - Martin Serchinger
- Department of Molecular Hematology, Goethe University Medical School, D-60590 Frankfurt am Main, Germany
| | - Frank Schnütgen
- Department of Molecular Hematology, Goethe University Medical School, D-60590 Frankfurt am Main, Germany
| | - Duran Sürün
- Department of Molecular Hematology, Goethe University Medical School, D-60590 Frankfurt am Main, Germany
| | - Anja Sterner-Kock
- Center for Experimental Medicine, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardiopulmonary System (ECCPS), Justus-Liebig-University Giessen, Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), D-35392 Giessen, Germany
| | - Harald von Melchner
- Department of Molecular Hematology, Goethe University Medical School, D-60590 Frankfurt am Main, Germany.
| |
Collapse
|
109
|
Sipilä KH, Ranga V, Rappu P, Torittu A, Pirilä L, Käpylä J, Johnson MS, Larjava H, Heino J. Extracellular citrullination inhibits the function of matrix associated TGF-β. Matrix Biol 2016; 55:77-89. [DOI: 10.1016/j.matbio.2016.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/18/2016] [Accepted: 02/21/2016] [Indexed: 10/22/2022]
|
110
|
Fang F, Marangoni RG, Zhou X, Yang Y, Ye B, Shangguang A, Qin W, Wang W, Bhattacharyya S, Wei J, Tourtellotte WG, Varga J. Toll-like Receptor 9 Signaling Is Augmented in Systemic Sclerosis and Elicits Transforming Growth Factor β-Dependent Fibroblast Activation. Arthritis Rheumatol 2016; 68:1989-2002. [PMID: 26946325 PMCID: PMC9993331 DOI: 10.1002/art.39655] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 02/18/2016] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Although transforming growth factor β (TGFβ) is recognized as being a key trigger of fibroblast activation in systemic sclerosis (SSc), prominent innate immunity suggests that additional pathways contribute to disease persistence. Toll-like receptor 9 (TLR9) is implicated in autoimmunity and fibrosis; however, the expression, mechanism of action, and pathogenic role of TLR9 signaling in SSc remain uncharacterized. The aim of this study was to explore the expression, activity, and potential pathogenic role of TLR9 in the context of skin fibrosis in SSc and in mouse models of experimental fibrosis. METHODS Expression and localization of TLR9 were evaluated in SSc skin biopsy specimens and explanted skin fibroblasts. Fibrotic responses elicited by type A CpG oligonucleotide and mitochondrial DNA (mtDNA) were examined in human skin fibroblasts by a combination of real-time quantitative polymerase chain reaction, Western blot analysis, transient transfection, immunofluorescence microscopy, and functional assays. Expression of TLR9 was examined in 2 distinct mouse models of experimental fibrosis. RESULTS Skin biopsy specimens obtained from 2 independent cohorts of SSc patients showed up-regulation of TLR9, and myofibroblasts were the major cellular source. Moreover, SSc skin biopsy specimens showed evidence of TLR9 pathway activation. CpG induced robust TLR9-dependent fibrotic responses in explanted normal fibroblasts that could be blocked by bortezomib and were mediated through the action of endogenous TGFβ. Mice with experimental fibrosis showed a time-dependent increase in TLR9 localized primarily to myofibroblasts in the dermis. CONCLUSION In isolated fibroblasts, TLR9 elicits fibrotic responses mediated via endogenous TGFβ. In patients with SSc, mtDNA and other damage-associated TLR9 ligands in the skin might trigger localized activation of TLR9 signaling, TGFβ production, and consequent fibroblast activation. Disrupting this fibrotic process with inhibitors targeting TLR9 or its downstream signaling pathways might therefore represent a novel approach to SSc therapy.
Collapse
Affiliation(s)
- Feng Fang
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | | | - Yang Yang
- China Pharmaceutical University, Nanjing, China
| | - Boping Ye
- China Pharmaceutical University, Nanjing, China
| | - Anna Shangguang
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Wenyi Qin
- University of Illinois at Chicago, Chicago, Illinois
| | - Wenxia Wang
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Jun Wei
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - John Varga
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
111
|
Wilbers RHP, Westerhof LB, van Raaij DR, van Adrichem M, Prakasa AD, Lozano-Torres JL, Bakker J, Smant G, Schots A. Co-expression of the protease furin in Nicotiana benthamiana leads to efficient processing of latent transforming growth factor-β1 into a biologically active protein. PLANT BIOTECHNOLOGY JOURNAL 2016; 14:1695-704. [PMID: 26834022 PMCID: PMC5067602 DOI: 10.1111/pbi.12530] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/16/2015] [Accepted: 10/19/2015] [Indexed: 05/21/2023]
Abstract
Transforming growth factor beta (TGF-β) is a signalling molecule that plays a key role in developmental and immunological processes in mammals. Three TGF-β isoforms exist in humans, and each isoform has unique therapeutic potential. Plants offer a platform for the production of recombinant proteins, which is cheap and easy to scale up and has a low risk of contamination with human pathogens. TGF-β3 has been produced in plants before using a chloroplast expression system. However, this strategy requires chemical refolding to obtain a biologically active protein. In this study, we investigated the possibility to transiently express active human TGF-β1 in Nicotiana benthamiana plants. We successfully expressed mature TGF-β1 in the absence of the latency-associated peptide (LAP) using different strategies, but the obtained proteins were inactive. Upon expression of LAP-TGF-β1, we were able to show that processing of the latent complex by a furin-like protease does not occur in planta. The use of a chitinase signal peptide enhanced the expression and secretion of LAP-TGF-β1, and co-expression of human furin enabled the proteolytic processing of latent TGF-β1. Engineering the plant post-translational machinery by co-expressing human furin also enhanced the accumulation of biologically active TGF-β1. This engineering step is quite remarkable, as furin requires multiple processing steps and correct localization within the secretory pathway to become active. Our data demonstrate that plants can be a suitable platform for the production of complex proteins that rely on specific proteolytic processing.
Collapse
Affiliation(s)
- Ruud H P Wilbers
- Laboratory of Nematology, Plant Sciences Department, Wageningen University and Research Centre, Wageningen, The Netherlands
| | - Lotte B Westerhof
- Laboratory of Nematology, Plant Sciences Department, Wageningen University and Research Centre, Wageningen, The Netherlands
| | - Debbie R van Raaij
- Laboratory of Nematology, Plant Sciences Department, Wageningen University and Research Centre, Wageningen, The Netherlands
| | - Marloes van Adrichem
- Laboratory of Nematology, Plant Sciences Department, Wageningen University and Research Centre, Wageningen, The Netherlands
| | - Andreas D Prakasa
- Laboratory of Nematology, Plant Sciences Department, Wageningen University and Research Centre, Wageningen, The Netherlands
| | - Jose L Lozano-Torres
- Laboratory of Nematology, Plant Sciences Department, Wageningen University and Research Centre, Wageningen, The Netherlands
| | - Jaap Bakker
- Laboratory of Nematology, Plant Sciences Department, Wageningen University and Research Centre, Wageningen, The Netherlands
| | - Geert Smant
- Laboratory of Nematology, Plant Sciences Department, Wageningen University and Research Centre, Wageningen, The Netherlands
| | - Arjen Schots
- Laboratory of Nematology, Plant Sciences Department, Wageningen University and Research Centre, Wageningen, The Netherlands
| |
Collapse
|
112
|
Jones K, Ballesteros A, Mentink-Kane M, Warren J, Rattila S, Malech H, Kang E, Dveksler G. PSG9 Stimulates Increase in FoxP3+ Regulatory T-Cells through the TGF-β1 Pathway. PLoS One 2016; 11:e0158050. [PMID: 27389696 PMCID: PMC4936685 DOI: 10.1371/journal.pone.0158050] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 06/09/2016] [Indexed: 12/20/2022] Open
Abstract
The pregnancy-specific glycoproteins (PSGs) are a family of proteins secreted by the syncytiotrophoblast of the placenta and are the most abundant trophoblastic proteins in maternal blood during the third trimester. The human PSG family consists of 10 protein-coding genes, some of which have a possible role in maintaining maternal immune tolerance to the fetus. PSG9 was reported as a potential predictive biomarker of pre-eclampsia, a serious complication of pregnancy that has been related to immunological dysfunction at the fetal-maternal interface. Therefore, we hypothesized that PSG9 may have an immunoregulatory role during pregnancy. We found that PSG9 binds to LAP and activates the latent form of TGF-β1. In addition, PSG9 induces the secretion of TGF-β1 from macrophages but not from CD4+ T-cells. TGF-β1 is required for the ex vivo differentiation of regulatory T-cells and, consistent with the ability of PSG9 to activate this cytokine, we observed that PSG9 induces the differentiation of FoxP3+ regulatory T-cells from naïve murine and human T-cells. Cytokines that are associated with inflammatory responses were also reduced in the supernatants of T-cells treated with PSG9, suggesting that PSG9, through its activation of TGFβ-1, could be a potent inducer of immune tolerance.
Collapse
Affiliation(s)
- Karlie Jones
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, United States of America
| | - Angela Ballesteros
- National Institute on Deafness and other Communication Disorders, NIH, Bethesda, Maryland, United States of America
| | | | - James Warren
- Department of Pathology, USUHS, Bethesda, Maryland, 20814, United States of America
| | - Shemona Rattila
- Department of Pathology, USUHS, Bethesda, Maryland, 20814, United States of America
| | - Harry Malech
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, United States of America
| | - Elizabeth Kang
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, United States of America
| | - Gabriela Dveksler
- Department of Pathology, USUHS, Bethesda, Maryland, 20814, United States of America
- * E-mail:
| |
Collapse
|
113
|
Ulmasov B, Neuschwander-Tetri BA, Lai J, Monastyrskiy V, Bhat T, Yates MP, Oliva J, Prinsen MJ, Ruminski PG, Griggs DW. Inhibitors of Arg-Gly-Asp-Binding Integrins Reduce Development of Pancreatic Fibrosis in Mice. Cell Mol Gastroenterol Hepatol 2016; 2:499-518. [PMID: 28174730 PMCID: PMC5042566 DOI: 10.1016/j.jcmgh.2016.03.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 03/04/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Pancreatic stellate cells (PSCs) regulate the development of chronic pancreatitis (CP) and are activated by the cytokine transforming growth factor β (TGFB). Integrins of the αv family promote TGFB signaling in mice, probably by interacting with the Arg-Gly-Asp (RGD) sequence of the TGFB latency-associated peptide, which frees TGFB to bind its cellular receptors. However, little is known about the role of integrins in the development of CP. We investigated the effects of small-molecule integrin inhibitors in a mouse model of CP. METHODS We induced CP in C57BL/6 female mice by repeated cerulein administration. An active RGD peptidomimetic compound (Center for World Health and Medicine [CWHM]-12) was delivered by continuous infusion, starting 3 days before or 5 days after cerulein administration began. Pancreata were collected and parenchymal atrophy, fibrosis, and activation of PSCs were assessed by histologic, gene, and protein expression analyses. We measured CWHM-12 effects on activation of TGFB in co-culture assays in which rat PSC cells (large T immortalized cells [LTC-14]) activate expression of a TGFB-sensitive promoter in reporter cells. RESULTS Pancreatic tissues of mice expressed messenger RNAs encoding subunits of RGD-binding integrins. Cerulein administration increased expression of these integrins, altered pancreatic cell morphology, and induced fibrosis. The integrin inhibitor CWHM-12 decreased acinar cell atrophy and loss, and substantially reduced fibrosis, activation of PSCs, and expression of genes regulated by TGFB. CWHM-12 also reduced established fibrosis in mice and blocked activation of TGFB in cultured cells. CONCLUSIONS Based on studies of a mouse model of CP and cultured PSCs, integrins that bind RGD sequences activate PSCs and promote the development of pancreatic fibrogenesis in mice. Small-molecule antagonists of this interaction might be developed for treatment of pancreatic fibrotic diseases.
Collapse
Key Words
- CP, chronic pancreatitis
- CTGF, connective tissue growth factor
- CWHM, Center for World Health and Medicine
- Col1a1, collagen type I α1
- DMEM, Dulbecco's modified Eagle medium
- DMSO, dimethyl sulfoxide
- ECM, extracellular matrix
- FBS, fetal bovine serum
- IC50, median inhibitory concentration
- Inflammation
- LAP, latency-associated peptide
- LTC-14, large T immortalized cells
- MLEC, mink lung epithelial cell
- MMP, matrix metallopeptidase
- PBS, phosphate-buffered saline
- PCR, polymerase chain reaction
- PSC, pancreatic stellate cell
- Pancreas
- Peptidomimetic
- RGD, arginine-glycine-aspartic acid
- Signal Transduction
- TGFB, transforming growth factor β
- mPSC, mouse pancreatic stellate cell
- mRNA, messenger RNA
- p-SMAD, phosphorylated SMAD
- α-SMA, α-smooth muscle actin
Collapse
Affiliation(s)
- Barbara Ulmasov
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Saint Louis University, Saint Louis, Missouri
| | - Brent A. Neuschwander-Tetri
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Saint Louis University, Saint Louis, Missouri
| | - Jinping Lai
- Department of Pathology, Saint Louis University, Saint Louis, Missouri
| | - Vladimir Monastyrskiy
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Saint Louis University, Saint Louis, Missouri
| | - Trisha Bhat
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Saint Louis University, Saint Louis, Missouri
| | - Matthew P. Yates
- Center for World Health and Medicine, Saint Louis University, Saint Louis, Missouri
| | - Jonathan Oliva
- Center for World Health and Medicine, Saint Louis University, Saint Louis, Missouri
| | - Michael J. Prinsen
- Center for World Health and Medicine, Saint Louis University, Saint Louis, Missouri
| | - Peter G. Ruminski
- Center for World Health and Medicine, Saint Louis University, Saint Louis, Missouri
| | - David W. Griggs
- Center for World Health and Medicine, Saint Louis University, Saint Louis, Missouri
| |
Collapse
|
114
|
Sasaki T, Hanisch FG, Deutzmann R, Sakai LY, Sakuma T, Miyamoto T, Yamamoto T, Hannappel E, Chu ML, Lanig H, von der Mark K. Functional consequence of fibulin-4 missense mutations associated with vascular and skeletal abnormalities and cutis laxa. Matrix Biol 2016; 56:132-149. [PMID: 27339457 DOI: 10.1016/j.matbio.2016.06.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/06/2016] [Accepted: 06/14/2016] [Indexed: 01/17/2023]
Abstract
Fibulin-4 is a 60kDa calcium binding glycoprotein that has an important role in development and integrity of extracellular matrices. It interacts with elastin, fibrillin-1 and collagen IV as well as with lysyl oxidases and is involved in elastogenesis and cross-link formation. To date, several mutations in the fibulin-4 gene (FBLN4/EFEMP2) are known in patients whose major symptoms are vascular deformities, aneurysm, cutis laxa, joint laxity, or arachnodactyly. The pathogenetic mechanisms how these mutations translate into the clinical phenotype are, however, poorly understood. In order to elucidate these mechanisms, we expressed fibulin-4 mutants recombinantly in HEK293 cells, purified the proteins in native forms and analyzed alterations in protein synthesis, secretion, matrix assembly, and interaction with other proteins in relation to wild type fibulin-4. Our studies show that different mutations affect these properties in multiple ways, resulting in fibulin-4 deficiency and/or impaired ability to form elastic fibers. The substitutions E126K and C267Y impaired secretion of the protein, but not mRNA synthesis. Furthermore, the E126K mutant showed less resistance to proteases, reduced binding to collagen IV and fibrillin-1, as well as to LTBP1s and LTBP4s. The A397T mutation introduced an extra O-glycosylation site and deleted binding to LTBP1s. We show that fibulin-4 binds stronger than fibulin-3 and -5 to LTBP1s, 3, and 4s, and to the lysyl oxidases LOX and LOXL1; the binding of fibulin-4 to the LOX propeptide was strongly reduced by the mutation E57K. These findings show that different mutations in the fibulin-4 gene result in different molecular defects affecting secretion rates, protein stability, LOX-induced cross-linking, or binding to other ECM components and molecules of the TGF-β pathway, and thus illustrate the complex role of fibulin-4 in connective tissue assembly.
Collapse
Affiliation(s)
- Takako Sasaki
- Department of Experimental Medicine I, Nikolaus-Fiebiger Center of Molecular Medicine, University of Erlangen-Nürnberg, 91054 Erlangen, Germany; Department of Biochemistry II, Faculty of Medicine, Oita University, Oita 879-5593, Japan.
| | - Franz-Georg Hanisch
- Institute for Biochemistry II, Medical Faculty, Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Rainer Deutzmann
- Institute of Biochemistry, Microbiology and Genetics, University of Regensburg, 93053 Regensburg, Germany
| | - Lynn Y Sakai
- Shriners Hospital for Children, Portland Research Center, Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Tetsushi Sakuma
- Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima 739-8526, Japan
| | - Tatsuo Miyamoto
- Department of Genetics and Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
| | - Takashi Yamamoto
- Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima 739-8526, Japan
| | - Ewald Hannappel
- Institut für Biochemie, Emil-Fischer-Zentrum, University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Mon-Li Chu
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Harald Lanig
- Central Institute for Scientific Computing (ZISC), University of Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Klaus von der Mark
- Department of Experimental Medicine I, Nikolaus-Fiebiger Center of Molecular Medicine, University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
115
|
Su P, Chen S, Zheng YH, Zhou HY, Yan CH, Yu F, Zhang YG, He L, Zhang Y, Wang Y, Wu L, Wu X, Yu B, Ma LY, Yang Z, Wang J, Zhao G, Zhu J, Wu ZY, Sun B. Novel Function of Extracellular Matrix Protein 1 in Suppressing Th17 Cell Development in Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2016; 197:1054-64. [PMID: 27316685 DOI: 10.4049/jimmunol.1502457] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 05/21/2016] [Indexed: 12/24/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the CNS characterized by demyelination and axonal damage. Experimental autoimmune encephalomyelitis (EAE) is a well-established animal model for human MS. Although Th17 cells are important for disease induction, Th2 cells are inhibitory in this process. In this article, we report the effect of a Th2 cell product, extracellular matrix protein 1 (ECM1), on the differentiation of Th17 cells and the development of EAE. Our results demonstrated that ECM1 administration from day 1 to day 7 following the EAE induction could ameliorate the Th17 cell responses and EAE development in vivo. Further study of the mechanism revealed that ECM1 could interact with αv integrin on dendritic cells and block the αv integrin-mediated activation of latent TGF-β, resulting in an inhibition of Th17 cell differentiation at an early stage of EAE induction. Furthermore, overexpression of ECM1 in vivo significantly inhibited the Th17 cell response and EAE induction in ECM1 transgenic mice. Overall, our work has identified a novel function of ECM1 in inhibiting Th17 cell differentiation in the EAE model, suggesting that ECM1 may have the potential to be used in clinical applications for understanding the pathogenesis of MS and its diagnosis.
Collapse
Affiliation(s)
- Pan Su
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Sheng Chen
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and the Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and Institute of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou 350004, China
| | - Yu Han Zheng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hai Yan Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Cheng Hua Yan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Fang Yu
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Ya Guang Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lan He
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuan Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yanming Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lei Wu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and the Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoai Wu
- Novo Nordisk Research Center, Beijing 100000, China
| | - Bingke Yu
- Novo Nordisk Research Center, Beijing 100000, China
| | - Li Yan Ma
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhiru Yang
- Novo Nordisk Research Center, Beijing 100000, China
| | - Jianhua Wang
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Guixian Zhao
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jinfang Zhu
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892;
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and the Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou 310058, China;
| | - Bing Sun
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China; and
| |
Collapse
|
116
|
Mohammed J, Beura LK, Bobr A, Astry B, Chicoine B, Kashem SW, Welty NE, Igyártó BZ, Wijeyesinghe S, Thompson EA, Matte C, Bartholin L, Kaplan A, Sheppard D, Bridges AG, Shlomchik WD, Masopust D, Kaplan DH. Stromal cells control the epithelial residence of DCs and memory T cells by regulated activation of TGF-β. Nat Immunol 2016; 17:414-21. [PMID: 26901152 PMCID: PMC5135085 DOI: 10.1038/ni.3396] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/05/2016] [Indexed: 01/02/2023]
Abstract
Cells of the immune system that reside in barrier epithelia provide a first line of defense against pathogens. Langerhans cells (LCs) and CD8(+) tissue-resident memory T cells (TRM cells) require active transforming growth factor-β1 (TGF-β) for epidermal residence. Here we found that integrins αvβ6 and αvβ8 were expressed in non-overlapping patterns by keratinocytes (KCs) and maintained the epidermal residence of LCs and TRM cells by activating latent TGF-β. Similarly, the residence of dendritic cells and TRM cells in the small intestine epithelium also required αvβ6. Treatment of the skin with ultraviolet irradiation decreased integrin expression on KCs and reduced the availability of active TGF-β, which resulted in LC migration. Our data demonstrated that regulated activation of TGF-β by stromal cells was able to directly control epithelial residence of cells of the immune system through a novel mechanism of intercellular communication.
Collapse
Affiliation(s)
- Javed Mohammed
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Lalit K Beura
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Aleh Bobr
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Brian Astry
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Brian Chicoine
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Sakeen W Kashem
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Nathan E Welty
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Botond Z Igyártó
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Sathi Wijeyesinghe
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Emily A Thompson
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Catherine Matte
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Laurent Bartholin
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Lyon, France
| | - Alesia Kaplan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Dean Sheppard
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Alina G Bridges
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Warren D Shlomchik
- Department of Medicine, University of Pittsburgh Cancer Center Institute, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh Cancer Center Institute, Pittsburgh, Pennsylvania, USA
| | - David Masopust
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Daniel H Kaplan
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
- Department of Dermatology, University of Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pennsylvania, USA
| |
Collapse
|
117
|
Luzina IG, Lockatell V, Hyun SW, Kopach P, Kang PH, Noor Z, Liu A, Lillehoj EP, Lee C, Miranda-Ribera A, Todd NW, Goldblum SE, Atamas SP. Elevated expression of NEU1 sialidase in idiopathic pulmonary fibrosis provokes pulmonary collagen deposition, lymphocytosis, and fibrosis. Am J Physiol Lung Cell Mol Physiol 2016; 310:L940-54. [PMID: 26993524 DOI: 10.1152/ajplung.00346.2015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 03/17/2016] [Indexed: 01/08/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) poses challenges to understanding its underlying cellular and molecular mechanisms and the development of better therapies. Previous studies suggest a pathophysiological role for neuraminidase 1 (NEU1), an enzyme that removes terminal sialic acid from glycoproteins. We observed increased NEU1 expression in epithelial and endothelial cells, as well as fibroblasts, in the lungs of patients with IPF compared with healthy control lungs. Recombinant adenovirus-mediated gene delivery of NEU1 to cultured primary human cells elicited profound changes in cellular phenotypes. Small airway epithelial cell migration was impaired in wounding assays, whereas, in pulmonary microvascular endothelial cells, NEU1 overexpression strongly impacted global gene expression, increased T cell adhesion to endothelial monolayers, and disrupted endothelial capillary-like tube formation. NEU1 overexpression in fibroblasts provoked increased levels of collagen types I and III, substantial changes in global gene expression, and accelerated degradation of matrix metalloproteinase-14. Intratracheal instillation of NEU1 encoding, but not control adenovirus, induced lymphocyte accumulation in bronchoalveolar lavage samples and lung tissues and elevations of pulmonary transforming growth factor-β and collagen. The lymphocytes were predominantly T cells, with CD8(+) cells exceeding CD4(+) cells by nearly twofold. These combined data indicate that elevated NEU1 expression alters functional activities of distinct lung cell types in vitro and recapitulates lymphocytic infiltration and collagen accumulation in vivo, consistent with mechanisms implicated in lung fibrosis.
Collapse
Affiliation(s)
- Irina G Luzina
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Virginia Lockatell
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Sang W Hyun
- University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Pavel Kopach
- University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Phillip H Kang
- University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Zahid Noor
- University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Anguo Liu
- University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Erik P Lillehoj
- University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Chunsik Lee
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | | | - Nevins W Todd
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Simeon E Goldblum
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Sergei P Atamas
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland; University of Maryland School of Medicine, Baltimore, Maryland; and
| |
Collapse
|
118
|
Sun KH, Chang Y, Reed NI, Sheppard D. α-Smooth muscle actin is an inconsistent marker of fibroblasts responsible for force-dependent TGFβ activation or collagen production across multiple models of organ fibrosis. Am J Physiol Lung Cell Mol Physiol 2016; 310:L824-36. [PMID: 26944089 DOI: 10.1152/ajplung.00350.2015] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 02/29/2016] [Indexed: 01/18/2023] Open
Abstract
Fibrosis is a common pathological sequela of tissue injury or inflammation, and is a major cause of organ failure. Subsets of fibroblasts contribute to tissue fibrosis in multiple ways, including generating contractile force to activate integrin-bound, latent TGFβ and secreting excess amounts of collagens and other extracellular matrix proteins (ECM) that make up pathologic scar. However, the precise fibroblast subsets that drive fibrosis have been poorly understood. In the absence of well-characterized markers, α-smooth muscle actin (αSMA) is often used to identify pathologic fibroblasts, and some authors have equated αSMA(+) cells with contractile myofibroblasts and proposed that these cells are the major source of ECM. Here, we investigated how well αSMA expression describes fibroblast subsets responsible for TGFβ activation and collagen production in three commonly used models of organ fibrosis that we previously reported could be inhibited by loss of αv integrins on all fibroblasts (using PDGFRβ-Cre). Interestingly, αSMA-directed deletion of αv integrins protected mice from CCl4-induced hepatic fibrosis, but not bleomycin-induced pulmonary or unilateral ureteral obstruction-induced renal fibrosis. Using Col-EGFP/αSMA-RFP dual reporter mice, we found that only a minority of collagen-producing cells coexpress αSMA in the fibrotic lung and kidney. Notably, Col-EGFP(+)αSMA-RFP(-) cells isolated from the fibrotic lung and kidney were equally capable of activating TGFβ as were Col-EGFP(+)αSMA-RFP(+) cells from the same organ, and this TGFβ activation was blocked by a TGFβ-blocking antibody and an inhibitor of nonmuscle myosin, respectively. Taken together, our results suggest that αSMA is an inconsistent marker of contractile and collagen-producing fibroblasts in murine experimental models of organ fibrosis.
Collapse
Affiliation(s)
- Kai-Hui Sun
- Lung Biology Center, Department of Medicine, University of California, San Francisco; and
| | - Yongen Chang
- Lung Biology Center, Department of Medicine, University of California, San Francisco; and Division of Nephrology, Department of Medicine, University of California, Irvine, Orange, California
| | - Nilgun I Reed
- Lung Biology Center, Department of Medicine, University of California, San Francisco; and
| | - Dean Sheppard
- Lung Biology Center, Department of Medicine, University of California, San Francisco; and
| |
Collapse
|
119
|
Flanagan-Steet H, Aarnio M, Kwan B, Guihard P, Petrey A, Haskins M, Blanchard F, Steet R. Cathepsin-Mediated Alterations in TGFß-Related Signaling Underlie Disrupted Cartilage and Bone Maturation Associated With Impaired Lysosomal Targeting. J Bone Miner Res 2016; 31:535-48. [PMID: 26404503 PMCID: PMC4808492 DOI: 10.1002/jbmr.2722] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 09/21/2015] [Accepted: 09/23/2015] [Indexed: 11/11/2022]
Abstract
Hypersecretion of acid hydrolases is a hallmark feature of mucolipidosis II (MLII), a lysosomal storage disease caused by loss of carbohydrate-dependent lysosomal targeting. Inappropriate extracellular action of these hydrolases is proposed to contribute to skeletal pathogenesis, but the mechanisms that connect hydrolase activity to the onset of disease phenotypes remain poorly understood. Here we link extracellular cathepsin K activity to abnormal bone and cartilage development in MLII animals by demonstrating that it disrupts the balance of TGFß-related signaling during chondrogenesis. TGFß-like Smad2,3 signals are elevated and BMP-like Smad1,5,8 signals reduced in both feline and zebrafish MLII chondrocytes and osteoblasts, maintaining these cells in an immature state. Reducing either cathepsin K activity or expression of the transcriptional regulator Sox9a in MLII zebrafish significantly improved phenotypes. We further identify components of the large latent TGFß complex as novel targets of cathepsin K at neutral pH, providing a possible mechanism for enhanced Smad2,3 activation in vivo. These findings highlight the complexity of the skeletal disease associated with MLII and bring new insight to the role of secreted cathepsin proteases in cartilage development and growth factor regulation.
Collapse
Affiliation(s)
| | - Megan Aarnio
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Brian Kwan
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | | | - Aaron Petrey
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Mark Haskins
- Departments of Pathology and Clinical Studies, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | | | - Richard Steet
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| |
Collapse
|
120
|
Reed NI, Jo H, Chen C, Tsujino K, Arnold TD, DeGrado WF, Sheppard D. The αvβ1 integrin plays a critical in vivo role in tissue fibrosis. Sci Transl Med 2016; 7:288ra79. [PMID: 25995225 DOI: 10.1126/scitranslmed.aaa5094] [Citation(s) in RCA: 234] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Integrins are transmembrane heterodimeric receptors that contribute to diverse biological functions and play critical roles in many human diseases. Studies using integrin subunit knockout mice and inhibitory antibodies have identified important roles for nearly every integrin heterodimer and led to the development of a number of potentially useful therapeutics. One notable exception is the αvβ1 integrin. αv and β1 subunits are individually present in numerous dimer pairs, making it challenging to infer specific roles for αvβ1 by genetic inactivation of individual subunits, and αvβ1 complex-specific blocking antibodies do not yet exist. We therefore developed a potent and highly specific small-molecule inhibitor of αvβ1 to probe the function of this understudied integrin. We found that αvβ1, which is highly expressed on activated fibroblasts, directly binds to the latency-associated peptide of transforming growth factor-β1 (TGFβ1) and mediates TGFβ1 activation. Therapeutic delivery of this αvβ1 inhibitor attenuated bleomycin-induced pulmonary fibrosis and carbon tetrachloride-induced liver fibrosis, suggesting that drugs based on this lead compound could be broadly useful for treatment of diseases characterized by excessive tissue fibrosis.
Collapse
Affiliation(s)
- Nilgun I Reed
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hyunil Jo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Chun Chen
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kazuyuki Tsujino
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Thomas D Arnold
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - William F DeGrado
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Dean Sheppard
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
121
|
Huang SS, Chen CL, Huang FW, Hou WH, Huang JS. DMSO Enhances TGF-β Activity by Recruiting the Type II TGF-β Receptor From Intracellular Vesicles to the Plasma Membrane. J Cell Biochem 2016; 117:1568-79. [PMID: 26587792 DOI: 10.1002/jcb.25448] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 11/18/2015] [Indexed: 01/03/2023]
Abstract
Dimethyl sulfoxide (DMSO) is used to treat many diseases/symptoms. The molecular basis of the pharmacological actions of DMSO has been unclear. We hypothesized that DMSO exerts some of these actions by enhancing TGF-β activity. Here we show that DMSO enhances TGF-β activity by ∼3-4-fold in Mv1Lu and NMuMG cells expressing Smad-dependent luciferase reporters. In Mv1Lu cells, DMSO enhances TGF-β-stimulated expression of P-Smad2 and PAI-1. It increases cell-surface expression of TGF-β receptors (TβR-I and/or TβR-II) by ∼3-4-fold without altering their cellular levels as determined by (125) I-labeled TGF-β-cross-linking/Western blot analysis, suggesting the presence of large intracellular pools in these cells. Sucrose density gradient ultracentrifugation/Western blot analysis reveals that DMSO induces recruitment of TβR-II (but not TβR-I) from its intracellular pool to plasma-membrane microdomains. It induces more recruitment of TβR-II to non-lipid raft microdomains than to lipid rafts/caveolae. Mv1Lu cells transiently transfected with TβR-II-HA plasmid were treated with DMSO and analyzed by indirect immunofluoresence staining using anti-HA antibody. In these cells, TβR-II-HA is present as a vesicle-like network in the cytoplasm as well as in the plasma membrane. DMSO causes depletion of TβR-II-HA-containing vesicles from the cytoplasm and co-localization of TβR-II-HA and cveolin-1 at the plasma membrane. These results suggest that DMSO, a fusogenic substance, enhances TGF-β activity presumably by inducing fusion of cytoplasmic vesicles (containing TβR-II) and the plasma membrane, resulting in increased localization of TβR-II to non-lipid raft microdomains where canonical signaling occurs. Fusogenic activity of DMSO may play a pivotal role in its pharmacological actions involving membrane proteins with large cytoplasmic pools. J. Cell. Biochem. 117: 1568-1579, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Chun-Lin Chen
- Department of Biological Science, National Sun Yat-sen University and Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, 804, Taiwan
| | - Franklin W Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston and Harvard Medical School, Boston, Massachusetts, 02115
| | - Wei-Hsien Hou
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Doisy Research Center, 1100 S. Grand Boulevard, St. Louis, Missouri, 63104
| | - Jung San Huang
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Doisy Research Center, 1100 S. Grand Boulevard, St. Louis, Missouri, 63104
| |
Collapse
|
122
|
West JD, Carrier EJ, Bloodworth NC, Schroer AK, Chen P, Ryzhova LM, Gladson S, Shay S, Hutcheson JD, Merryman WD. Serotonin 2B Receptor Antagonism Prevents Heritable Pulmonary Arterial Hypertension. PLoS One 2016; 11:e0148657. [PMID: 26863209 PMCID: PMC4749293 DOI: 10.1371/journal.pone.0148657] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 01/21/2016] [Indexed: 12/21/2022] Open
Abstract
Serotonergic anorexigens are the primary pharmacologic risk factor associated with pulmonary arterial hypertension (PAH), and the resulting PAH is clinically indistinguishable from the heritable form of disease, associated with BMPR2 mutations. Both BMPR2 mutation and agonists to the serotonin receptor HTR2B have been shown to cause activation of SRC tyrosine kinase; conversely, antagonists to HTR2B inhibit SRC trafficking and downstream function. To test the hypothesis that a HTR2B antagonist can prevent BMRP2 mutation induced PAH by restricting aberrant SRC trafficking and downstream activity, we exposed BMPR2 mutant mice, which spontaneously develop PAH, to a HTR2B antagonist, SB204741, to block the SRC activation caused by BMPR2 mutation. SB204741 prevented the development of PAH in BMPR2 mutant mice, reduced recruitment of inflammatory cells to their lungs, and reduced muscularization of their blood vessels. By atomic force microscopy, we determined that BMPR2 mutant mice normally had a doubling of vessel stiffness, which was substantially normalized by HTR2B inhibition. SB204741 reduced SRC phosphorylation and downstream activity in BMPR2 mutant mice. Gene expression arrays indicate that the primary changes were in cytoskeletal and muscle contractility genes. These results were confirmed by gel contraction assays showing that HTR2B inhibition nearly normalizes the 400% increase in gel contraction normally seen in BMPR2 mutant smooth muscle cells. Heritable PAH results from increased SRC activation, cellular contraction, and vascular resistance, but antagonism of HTR2B prevents SRC phosphorylation, downstream activity, and PAH in BMPR2 mutant mice.
Collapse
MESH Headings
- Animals
- Bone Morphogenetic Protein Receptors, Type II/deficiency
- Bone Morphogenetic Protein Receptors, Type II/genetics
- Cell Movement/drug effects
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Gene Expression Profiling
- Gene Expression Regulation
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/prevention & control
- Indoles/pharmacology
- Lung/drug effects
- Lung/metabolism
- Lung/pathology
- Mice
- Mice, Transgenic
- Muscle Contraction/drug effects
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Mutation
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Oligonucleotide Array Sequence Analysis
- Phosphorylation
- Protein Transport
- Receptor, Serotonin, 5-HT2B/genetics
- Receptor, Serotonin, 5-HT2B/metabolism
- Serotonin Antagonists/pharmacology
- Signal Transduction
- Urea/analogs & derivatives
- Urea/pharmacology
- Vascular Stiffness/drug effects
- src-Family Kinases/antagonists & inhibitors
- src-Family Kinases/genetics
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- James D. West
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, United States of America
- * E-mail: (JDW); (WDM)
| | - Erica J. Carrier
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, United States of America
| | - Nathaniel C. Bloodworth
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37232, United States of America
| | - Alison K. Schroer
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37232, United States of America
| | - Peter Chen
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, United States of America
| | - Larisa M. Ryzhova
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37232, United States of America
| | - Santhi Gladson
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, United States of America
| | - Sheila Shay
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, United States of America
| | - Joshua D. Hutcheson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37232, United States of America
| | - W. David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37232, United States of America
- * E-mail: (JDW); (WDM)
| |
Collapse
|
123
|
Wang W, Burg N, Vootukuri S, Coller BS. Increased Smad2/3 phosphorylation in circulating leukocytes and platelet-leukocyte aggregates in a mouse model of aortic valve stenosis: Evidence of systemic activation of platelet-derived TGF-β1 and correlation with cardiac dysfunction. Blood Cells Mol Dis 2016; 58:1-5. [PMID: 27067480 DOI: 10.1016/j.bcmd.2016.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 01/21/2016] [Indexed: 10/22/2022]
Abstract
BACKGROUND Transforming growth factor-β1 (TGF-β1) has been implicated in the pathogenesis of aortic valve stenosis (AS). There is, however, little direct evidence for a role of active TGF-β1 in AS due to the sensitivity of current assays. We searched for evidence of plasma TGF-β1 activation by assaying Smad2/3 phosphorylation in circulating leukocytes and platelet-leukocyte aggregates (PLAs) in a mouse model of AS (Reversa). METHODS Echocardiography was used to measure AS and cardiac function. Intracellular phospho-flow cytometry in combination with optical fluorescence microscopy was used to detect PLAs and p-Smad2/3 levels. RESULTS Reversa mice on a western diet developed AS, had significantly increased numbers of PLAs and more intense staining for p-Smad2/3 in both PLAs and single leukocytes (all p<0.05). p-Smad2/3 staining was more intense in PLAs than in single leukocytes in both diet groups (p<0.05) and correlated with plasma total TGF-β1 levels (r=0.38, p=0.05 for PLAs and r=0.37, p=0.06 for single leukocytes) and reductions in ejection fraction (r=-0.42, p=0.03 for PLAs and r=-0.37, p=0.06 for single leukocytes). CONCLUSIONS p-Smad2/3 staining is more intense in leukocytes of hypercholesterolemic mice that developed AS, suggesting increased circulating active TGF-β1 levels. Leukocyte p-Smad2/3 may be a valuable surrogate indicator of circulating active TGF-β1.
Collapse
Affiliation(s)
- Wei Wang
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, NY, United States
| | - Nathalie Burg
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, NY, United States
| | - Spandana Vootukuri
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, NY, United States
| | - Barry S Coller
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, NY, United States.
| |
Collapse
|
124
|
Sun L, Dutta RK, Xie P, Kanwar YS. myo-Inositol Oxygenase Overexpression Accentuates Generation of Reactive Oxygen Species and Exacerbates Cellular Injury following High Glucose Ambience: A NEW MECHANISM RELEVANT TO THE PATHOGENESIS OF DIABETIC NEPHROPATHY. J Biol Chem 2016; 291:5688-5707. [PMID: 26792859 DOI: 10.1074/jbc.m115.669952] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Indexed: 01/23/2023] Open
Abstract
Diabetic nephropathy (DN) is characterized by perturbations in metabolic/cellular signaling pathways with generation of reactive oxygen species (ROS). The ROS are regarded as a common denominator of various pathways, and they inflict injury on renal glomerular cells. Recent studies indicate that tubular pathobiology also plays a role in the progression of DN. However, the mechanism(s) for how high (25 mm) glucose (HG) ambience induces tubular damage remains enigmatic. myo-Inositol oxygenase (MIOX) is a tubular enzyme that catabolizes myo-inositol to d-glucuronate via the glucuronate-xylulose (G-X) pathway. In this study, we demonstrated that G-X pathway enzymes are expressed in the kidney, and MIOX expression/bioactivity was up-regulated under HG ambience in LLC-PK1 cells, a tubular cell line. We further investigated whether MIOX overexpression leads to accentuation of tubulo-interstitial injury, as gauged by some of the parameters relevant to the progression of DN. Under HG ambience, MIOX overexpression accentuated redox imbalance, perturbed NAD(+)/NADH ratios, increased ROS generation, depleted reduced glutathione, reduced GSH/GSSG ratio, and enhanced adaptive changes in the profile of the antioxidant defense system. These changes were also accompanied by mitochondrial dysfunctions, DNA damage and induction of apoptosis, accentuated activity of profibrogenic cytokine, and expression of fibronectin, the latter two being the major hallmarks of DN. These perturbations were largely blocked by various ROS inhibitors (Mito Q, diphenyleneiodonium chloride, and N-acetylcysteine) and MIOX/NOX4 siRNA. In conclusion, this study highlights a novel mechanism where MIOX under HG ambience exacerbates renal injury during the progression of diabetic nephropathy following the generation of excessive ROS via an unexplored G-X pathway.
Collapse
Affiliation(s)
- Lin Sun
- From the Department of Nephrology and Renal Institute, 2nd Xiangya Hospital, Central South University, Changsha, Hunan 410011, China and
| | - Rajesh K Dutta
- the Departments of Pathology and Medicine, Northwestern University, Chicago, Illinois 60611
| | - Ping Xie
- the Departments of Pathology and Medicine, Northwestern University, Chicago, Illinois 60611
| | - Yashpal S Kanwar
- the Departments of Pathology and Medicine, Northwestern University, Chicago, Illinois 60611.
| |
Collapse
|
125
|
Cheng H, Chen T, Tor M, Park D, Zhou Q, Huang JB, Khatib N, Rong L, Zhou G. A High-Throughput Screening Platform Targeting PDLIM5 for Pulmonary Hypertension. ACTA ACUST UNITED AC 2016; 21:333-41. [DOI: 10.1177/1087057115625924] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/16/2015] [Indexed: 11/16/2022]
Abstract
Pulmonary arterial hypertension is a complex disease with multiple etiologic factors. PDLIM5, a member of the Enigma subfamily of PDZ and LIM domain protein family, contains an N-terminal PDZ domain and three LIM domains at its C-terminus. We have previously shown that overexpression of PDLIM5 prevents hypoxia-induced pulmonary hypertension (PH), and deletion of PDLIM5 in smooth muscle cells enhances hypoxia-induced PH in vivo. These results suggest that PDLIM5 may be a novel therapeutic target of PH. In this study, we aim to establish a high-throughput screening platform for PDLIM5-targeted drug discovery. We generated a stable mink lung epithelial cell line (MLEC) containing a transforming growth factor–β/Smad luciferase reporter with lentivirus-mediated suppression of PDLIM5 (MLEC-shPDLIM5) and measured levels of Smad2/3 and pSmad2/3. We found that in MLEC, suppression of PDLIM5 decreased Smad-dependent luciferase activity, Smad3, and pSmad3. We used MLEC-shPDLIM5 and a control cell line (MLEC-shCTL) to screen the Prestwick library (1200 compounds) and identified and validated paclitaxel as a PDLIM5 inhibitor in MLEC. Furthermore, we showed that paclitaxel inhibited Smad2 expression and Smad3 phosphorylation in A549 cells. Our study suggests that this system is robust and suitable for PDLIM5-targeted drug discovery.
Collapse
Affiliation(s)
- Han Cheng
- Department of Microbiology and Immunology, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Tianji Chen
- Department of Pediatrics, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Merve Tor
- Department of Pediatrics, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Deborah Park
- Department of Pediatrics, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Qiyuan Zhou
- Department of Pediatrics, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Jason B. Huang
- Department of Pediatrics, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Nour Khatib
- Department of Pediatrics, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Guofei Zhou
- Department of Pediatrics, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
- University of Illinois Cancer Center, Chicago, IL, USA
| |
Collapse
|
126
|
Smaldone S, Clayton NP, del Solar M, Pascual G, Cheng SH, Wentworth BM, Schaffler MB, Ramirez F. Fibrillin-1 Regulates Skeletal Stem Cell Differentiation by Modulating TGFβ Activity Within the Marrow Niche. J Bone Miner Res 2016; 31:86-97. [PMID: 26189658 PMCID: PMC5776390 DOI: 10.1002/jbmr.2598] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 07/07/2015] [Accepted: 07/16/2015] [Indexed: 12/17/2022]
Abstract
A full understanding of the microenvironmental factors that control the activities of skeletal stem cells (also known as mesenchymal stem cells [MSCs]) in the adult bone marrow holds great promise for developing new therapeutic strategies to mitigate age-related diseases of bone and cartilage degeneration. Bone loss is an understudied manifestation of Marfan syndrome, a multisystem disease associated with mutations in the extracellular matrix protein and TGFβ modulator fibrillin-1. Here we demonstrate that progressive loss of cancellous bone in mice with limbs deficient for fibrillin-1 (Fbn1(Prx1-/-) mice) is accounted for by premature depletion of MSCs and osteoprogenitor cells combined with constitutively enhanced bone resorption. Longitudinal analyses of Fbn1(Prx1-/-) mice showed incremental bone loss and trabecular microarchitecture degeneration accompanied by a progressive decrease in the number and clonogenic potential of MSCs. Significant paucity of marrow fat cells in the long bones of Fbn1(Prx1-/-) mice, together with reduced adipogenic potential of marrow stromal cell cultures, indicated an additional defect in MSC differentiation. This postulate was corroborated by showing that an Fbn1-silenced osteoprogenitor cell line cultured in the presence of insulin yielded fewer than normal adipocytes and exhibited relatively lower PPARγ levels. Consonant with fibrillin-1 modulation of TGFβ bioavailability, cultures of marrow stromal cells from Fbn1(Prx1-/-) limb bones showed improper overactivation of latent TGFβ. In line with this finding, systemic TGFβ neutralization improved bone mass and trabecular microarchitecture along with normalizing the number of MSCs, osteoprogenitor cells, and marrow adipocytes. Collectively, our findings show that fibrillin-1 regulates MSC activity by modulating TGFβ bioavailability within the microenvironment of marrow niches.
Collapse
Affiliation(s)
- Silvia Smaldone
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Maria del Solar
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gemma Pascual
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | | | | | - Mitchell B Schaffler
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
- Department of Biomedical Engineering, City College of New York, New York, NY, USA
| | - Francesco Ramirez
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
127
|
Muthusamy BP, Budi EH, Katsuno Y, Lee MK, Smith SM, Mirza AM, Akhurst RJ, Derynck R. ShcA Protects against Epithelial-Mesenchymal Transition through Compartmentalized Inhibition of TGF-β-Induced Smad Activation. PLoS Biol 2015; 13:e1002325. [PMID: 26680585 PMCID: PMC4682977 DOI: 10.1371/journal.pbio.1002325] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/10/2015] [Indexed: 12/15/2022] Open
Abstract
Epithelial–mesenchymal transition (EMT) is a normal cell differentiation event during development and contributes pathologically to carcinoma and fibrosis progression. EMT often associates with increased transforming growth factor-β (TGF-β) signaling, and TGF-β drives EMT, in part through Smad-mediated reprogramming of gene expression. TGF-β also activates the Erk MAPK pathway through recruitment and Tyr phosphorylation of the adaptor protein ShcA by the activated TGF-β type I receptor. We found that ShcA protects the epithelial integrity of nontransformed cells against EMT by repressing TGF-β-induced, Smad-mediated gene expression. p52ShcA competed with Smad3 for TGF-β receptor binding, and down-regulation of ShcA expression enhanced autocrine TGF-β/Smad signaling and target gene expression, whereas increased p52ShcA expression resulted in decreased Smad3 binding to the TGF-β receptor, decreased Smad3 activation, and increased Erk MAPK and Akt signaling. Furthermore, p52ShcA sequestered TGF-β receptor complexes to caveolin-associated membrane compartments, and reducing ShcA expression enhanced the receptor localization in clathrin-associated membrane compartments that enable Smad activation. Consequently, silencing ShcA expression induced EMT, with increased cell migration, invasion, and dissemination, and increased stem cell generation and mammosphere formation, dependent upon autocrine TGF-β signaling. These findings position ShcA as a determinant of the epithelial phenotype by repressing TGF-β-induced Smad activation through differential partitioning of receptor complexes at the cell surface. The adaptor protein ShcA protects epithelial cells from transitioning toward a mesenchymal phenotype by controlling partitioning of the TGF-β receptor and repressing downstream Smad2/3 activation. TGF-β family proteins control cell differentiation and various cell functions. Increased TGF-β signaling, acting through heteromeric receptor complexes, contributes to carcinoma progression and fibrosis. TGF-β drives epithelial–mesenchymal transdifferentiation (EMT), which enables cell migration and invasion. Upon TGF-β binding, “type I” receptors activate, through phosphorylation, Smad2 and Smad3 that control target gene transcription. In EMT, Smad complexes activate the expression of EMT “master” transcription factors and cooperate with these to repress the epithelial phenotype and activate mesenchymal gene expression. TGF-β receptors also activate Erk MAPK signaling, involving association of the adaptor protein ShcA and Tyr phosphorylation of ShcA by type I receptors. We now show that the predominant ShcA isoform, p52ShcA, competes with Smad2/3 for binding to type I TGF-β receptors, thus repressing Smad2/3 activation in response to TGF-β and localizing the receptors to caveolar compartments. Consequently, decreased ShcA expression enhanced TGF-β receptor localization in clathrin compartments and autocrine Smad2/3 signaling, repressed the epithelial phenotype, and promoted EMT. The changes following decreased ShcA expression resulted in increased cell migration and invasion, as well as increased stem cell generation, dependent upon autocrine TGF-β signaling. These findings position ShcA as a determinant of the epithelial phenotype by repressing TGF-β-induced Smad activation through differential partitioning of receptor complexes at the cell surface.
Collapse
Affiliation(s)
- Baby Periyanayaki Muthusamy
- Departments of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
| | - Erine H. Budi
- Departments of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
| | - Yoko Katsuno
- Departments of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
| | - Matthew K. Lee
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Susan M. Smith
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Amer M. Mirza
- XOMA Corp., Berkeley, California, United States of America
| | - Rosemary J. Akhurst
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
- Department of Anatomy, University of California, San Francisco, San Francisco, California, United States of America
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California, United States of America
| | - Rik Derynck
- Departments of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
- Department of Anatomy, University of California, San Francisco, San Francisco, California, United States of America
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
128
|
Friedrich U, Datta S, Schubert T, Plössl K, Schneider M, Grassmann F, Fuchshofer R, Tiefenbach KJ, Längst G, Weber BHF. Synonymous variants in HTRA1 implicated in AMD susceptibility impair its capacity to regulate TGF-β signaling. Hum Mol Genet 2015; 24:6361-73. [PMID: 26310622 DOI: 10.1093/hmg/ddv346] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 08/19/2015] [Indexed: 12/16/2023] Open
Abstract
High-temperature requirement A1 (HTRA1) is a secreted serine protease reported to play a role in the development of several cancers and neurodegenerative diseases. Still, the mechanism underlying the disease processes largely remains undetermined. In age-related macular degeneration (AMD), a common cause of vision impairment and blindness in industrialized societies, two synonymous polymorphisms (rs1049331:C>T, and rs2293870:G>T) in exon 1 of the HTRA1 gene were associated with a high risk to develop disease. Here, we show that the two polymorphisms result in a protein with altered thermophoretic properties upon heat-induced unfolding, trypsin accessibility and secretion behavior, suggesting unique structural features of the AMD-risk-associated HTRA1 protein. Applying MicroScale Thermophoresis and protease digestion analysis, we demonstrate direct binding and proteolysis of transforming growth factor β1 (TGF-β1) by normal HTRA1 but not the AMD-risk-associated isoform. As a consequence, both HTRA1 isoforms strongly differed in their ability to control TGF-β mediated signaling, as revealed by reporter assays targeting the TGF-β1-induced serpin peptidase inhibitor (SERPINE1, alias PAI-1) promoter. In addition, structurally altered HTRA1 led to an impaired autocrine TGF-β signaling in microglia, as measured by a strong down-regulation of downstream effectors of the TGF-β cascade such as phosphorylated SMAD2 and PAI-1 expression. Taken together, our findings demonstrate the effects of two synonymous HTRA1 variants on protein structure and protein interaction with TGF-β1. As a consequence, this leads to an impairment of TGF-β signaling and microglial regulation. Functional implications of the altered properties on AMD pathogenesis remain to be clarified.
Collapse
Affiliation(s)
- Ulrike Friedrich
- Institute of Human Genetics, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Shyamtanu Datta
- Institute of Human Genetics, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Thomas Schubert
- Department of Biochemistry, University of Regensburg, 2bind GmbH, Josef Engert Straße 13, 93053 Regensburg, Germany
| | - Karolina Plössl
- Institute of Human Genetics, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | | | - Felix Grassmann
- Institute of Human Genetics, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | | | - Klaus-Jürgen Tiefenbach
- Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany and
| | - Gernot Längst
- Department of Biochemistry, University of Regensburg
| | - Bernhard H F Weber
- Institute of Human Genetics, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany,
| |
Collapse
|
129
|
Iwasaki A, Sakai K, Moriya K, Sasaki T, Keene DR, Akhtar R, Miyazono T, Yasumura S, Watanabe M, Morishita S, Sakai T. Molecular Mechanism Responsible for Fibronectin-controlled Alterations in Matrix Stiffness in Advanced Chronic Liver Fibrogenesis. J Biol Chem 2015; 291:72-88. [PMID: 26553870 DOI: 10.1074/jbc.m115.691519] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Indexed: 12/23/2022] Open
Abstract
Fibrosis is characterized by extracellular matrix (ECM) remodeling and stiffening. However, the functional contribution of tissue stiffening to noncancer pathogenesis remains largely unknown. Fibronectin (Fn) is an ECM glycoprotein substantially expressed during tissue repair. Here we show in advanced chronic liver fibrogenesis using a mouse model lacking Fn that, unexpectedly, Fn-null livers lead to more extensive liver cirrhosis, which is accompanied by increased liver matrix stiffness and deteriorated hepatic functions. Furthermore, Fn-null livers exhibit more myofibroblast phenotypes and accumulate highly disorganized/diffuse collagenous ECM networks composed of thinner and significantly increased number of collagen fibrils during advanced chronic liver damage. Mechanistically, mutant livers show elevated local TGF-β activity and lysyl oxidase expressions. A significant amount of active lysyl oxidase is released in Fn-null hepatic stellate cells in response to TGF-β1 through canonical and noncanonical Smad such as PI3 kinase-mediated pathways. TGF-β1-induced collagen fibril stiffness in Fn-null hepatic stellate cells is significantly higher compared with wild-type cells. Inhibition of lysyl oxidase significantly reduces collagen fibril stiffness, and treatment of Fn recovers collagen fibril stiffness to wild-type levels. Thus, our findings indicate an indispensable role for Fn in chronic liver fibrosis/cirrhosis in negatively regulating TGF-β bioavailability, which in turn modulates ECM remodeling and stiffening and consequently preserves adult organ functions. Furthermore, this regulatory mechanism by Fn could be translated for a potential therapeutic target in a broader variety of chronic fibrotic diseases.
Collapse
Affiliation(s)
- Ayumi Iwasaki
- From the MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE, United Kingdom, the Graduate School of Biomedical Engineering and
| | - Keiko Sakai
- From the MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE, United Kingdom, the Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Kei Moriya
- the Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Takako Sasaki
- the Department of Biochemistry, Faculty of Medicine, Oita University, Oita, 879-5593, Japan
| | - Douglas R Keene
- the Micro-Imaging Center, Shriners Hospital for Children, Portland, Oregon 97231
| | - Riaz Akhtar
- Centre for Materials and Structures, School of Engineering, University of Liverpool, Liverpool L69 3GH, United Kingdom
| | - Takayoshi Miyazono
- Department of Gastroenterology and Hepatology, and Transfusion Medicine and Cell Therapy, Toyama University, Toyama 930-0194, Japan, and
| | - Satoshi Yasumura
- Department of Gastroenterology and Hepatology, and Transfusion Medicine and Cell Therapy, Toyama University, Toyama 930-0194, Japan, and
| | | | - Shin Morishita
- Environmental and Information Science, Yokohama National University, Yokohama 240-8501, Japan
| | - Takao Sakai
- From the MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE, United Kingdom, the Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195,
| |
Collapse
|
130
|
Talior-Volodarsky I, Arora PD, Wang Y, Zeltz C, Connelly KA, Gullberg D, McCulloch CA. Glycated Collagen Induces α11 Integrin Expression Through TGF-β2 and Smad3. J Cell Physiol 2015; 230:327-36. [PMID: 24962729 DOI: 10.1002/jcp.24708] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 06/20/2014] [Indexed: 01/21/2023]
Abstract
The adhesion of cardiac fibroblasts to the glycated collagen interstitium in diabetics is associated with de novo expression of the α11 integrin, myofibroblast formation and cardiac fibrosis. We examined how methylglyoxal-glycated collagen regulates α11 integrin expression. In cardiac fibroblasts plated on glycated collagen but not glycated fibronectin, there was markedly increased α11 integrin and α-smooth muscle actin expression. Compared with native collagen, binding of purified α11β1 integrin to glycated collagen was reduced by >fourfold, which was consistent with reduced fibroblast attachment to glycated collagen. Glycated collagen strongly enhanced the expression of TGF-β2 but not TGF-β1 or TGF-β3. The increased expression of TGF-β2 was inhibited by triple helical collagen peptides that mimic the α11β1 integrin binding site on type I collagen. In cardiac fibroblasts transfected with α11 integrin luciferase promoter constructs, glycated collagen activated the α11 integrin promoter. Analysis of α11 integrin promoter truncation mutants showed a novel Smad2/3 binding site located between -809 and -1300 nt that was required for promoter activation. We conclude that glycated collagen in the cardiac interstitium triggers an autocrine TGF-β2 signaling pathway that stimulates α11 integrin expression through Smad2/3 binding elements in the α11 integrin promoter, which is important for myofibroblast formation and fibrosis.
Collapse
Affiliation(s)
| | - Pamma D Arora
- Matrix Dynamics Group, University of Toronto, Toronto, Ontario, Canada
| | - Yongqiang Wang
- Matrix Dynamics Group, University of Toronto, Toronto, Ontario, Canada
| | - Cédric Zeltz
- Department of Biomedicine and Centre of Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, Bergen, Norway
| | - Kim A Connelly
- Keenan Research Centre for Biomedical Science of St. Michael Hospital, Toronto, Ontario, Canada
| | - Donald Gullberg
- Department of Biomedicine and Centre of Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, Bergen, Norway
| | | |
Collapse
|
131
|
Koistinen H, Hautala L, Koli K, Stenman UH. Absence of TGF-β Receptor Activation by Highly Purified hCG Preparations. Mol Endocrinol 2015; 29:1787-91. [PMID: 26495869 DOI: 10.1210/me.2015-1187] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Recently, several LH/human chorionic gonadotropin (hCG) receptor-independent activities for hCG have been described, including activation of the TGF-β receptor (TGFβR) by hyperglycosylated hCG and stimulation of trophoblast invasion. Because the hCG concentrations used in these studies have been rather high, reflecting physiological hCG levels in pregnancy, even a minor contamination with growth factors, which act at very low concentrations, may be significant. Several commercial hCG preparations have been found to contain significant amounts of epidermal growth factor (EGF), which we also confirmed here. Furthermore, we found that some hCG preparations also contain significant amounts of TGF-β1. These hCG preparations were able to activate ERK1/2 in JEG-3 choriocarcinoma cells or TGFβR in mink lung epithelial cells transfected with a reporter gene for TGFβR activation. No such activation was found with highly purified hCG or its free β-subunit (hCGβ), irrespective of whether they were hyperglycosylated or not. Taken together, our results suggest that the growth factor contaminations in the hCG preparations can cause activation of TGFβR and, at least in JEG-3 cells, MAPK signaling. This highlights the importance to carefully control for potential contaminations and that highly purified hCG preparations have to be used for biological studies.
Collapse
Affiliation(s)
- Hannu Koistinen
- Department of Clinical Chemistry (H.K., L.H., U.-H.S.), University of Helsinki and Helsinki University Central Hospital, 00290 Helsinki, Finland; and Research Programs Unit (K.K.), Translational Cancer Biology and Transplantation Laboratory, Haartman Institute, University of Helsinki, 00290 Helsinki, Finland
| | - Laura Hautala
- Department of Clinical Chemistry (H.K., L.H., U.-H.S.), University of Helsinki and Helsinki University Central Hospital, 00290 Helsinki, Finland; and Research Programs Unit (K.K.), Translational Cancer Biology and Transplantation Laboratory, Haartman Institute, University of Helsinki, 00290 Helsinki, Finland
| | - Katri Koli
- Department of Clinical Chemistry (H.K., L.H., U.-H.S.), University of Helsinki and Helsinki University Central Hospital, 00290 Helsinki, Finland; and Research Programs Unit (K.K.), Translational Cancer Biology and Transplantation Laboratory, Haartman Institute, University of Helsinki, 00290 Helsinki, Finland
| | - Ulf-Håkan Stenman
- Department of Clinical Chemistry (H.K., L.H., U.-H.S.), University of Helsinki and Helsinki University Central Hospital, 00290 Helsinki, Finland; and Research Programs Unit (K.K.), Translational Cancer Biology and Transplantation Laboratory, Haartman Institute, University of Helsinki, 00290 Helsinki, Finland
| |
Collapse
|
132
|
Huang SS, Chen CL, Huang FW, Johnson FE, Huang JS. Ethanol Enhances TGF-β Activity by Recruiting TGF-β Receptors From Intracellular Vesicles/Lipid Rafts/Caveolae to Non-Lipid Raft Microdomains. J Cell Biochem 2015; 117:860-71. [PMID: 26419316 DOI: 10.1002/jcb.25389] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 09/28/2015] [Indexed: 12/16/2022]
Abstract
Regular consumption of moderate amounts of ethanol has important health benefits on atherosclerotic cardiovascular disease (ASCVD). Overindulgence can cause many diseases, particularly alcoholic liver disease (ALD). The mechanisms by which ethanol causes both beneficial and harmful effects on human health are poorly understood. Here we demonstrate that ethanol enhances TGF-β-stimulated luciferase activity with a maximum of 0.5-1% (v/v) in Mv1Lu cells stably expressing a luciferase reporter gene containing Smad2-dependent elements. In Mv1Lu cells, 0.5% ethanol increases the level of P-Smad2, a canonical TGF-β signaling sensor, by ∼ 2-3-fold. Ethanol (0.5%) increases cell-surface expression of the type II TGF-β receptor (TβR-II) by ∼ 2-3-fold from its intracellular pool, as determined by I(125) -TGF-β-cross-linking/Western blot analysis. Sucrose density gradient ultracentrifugation and indirect immunofluorescence staining analyses reveal that ethanol (0.5% and 1%) also displaces cell-surface TβR-I and TβR-II from lipid rafts/caveolae and facilitates translocation of these receptors to non-lipid raft microdomains where canonical signaling occurs. These results suggest that ethanol enhances canonical TGF-β signaling by increasing non-lipid raft microdomain localization of the TGF-β receptors. Since TGF-β plays a protective role in ASCVD but can also cause ALD, the TGF-β enhancer activity of ethanol at low and high doses appears to be responsible for both beneficial and harmful effects. Ethanol also disrupts the location of lipid raft/caveolae of other membrane proteins (e.g., neurotransmitter, growth factor/cytokine, and G protein-coupled receptors) which utilize lipid rafts/caveolae as signaling platforms. Displacement of these membrane proteins induced by ethanol may result in a variety of pathologies in nerve, heart and other tissues.
Collapse
Affiliation(s)
| | - Chun-Lin Chen
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung, 804, Taiwan.,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, 804, Taiwan
| | - Franklin W Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, 02115.,Harvard Medical School, Boston, Massachusetts, 02115
| | - Frank E Johnson
- Department of Surgery, Saint Louis University School of Medicine, St. Louis, Missouri, 63104
| | - Jung San Huang
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Doisy Research Center, St. Louis, Missouri, 63104
| |
Collapse
|
133
|
Ekwueme EC, Shah JV, Mohiuddin M, Ghebes CA, Crispim JF, Saris DBF, Fernandes HAM, Freeman JW. Cross-Talk Between Human Tenocytes and Bone Marrow Stromal Cells Potentiates Extracellular Matrix Remodeling In Vitro. J Cell Biochem 2015; 117:684-93. [PMID: 26308651 DOI: 10.1002/jcb.25353] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 08/24/2015] [Indexed: 12/26/2022]
Abstract
Tendon and ligament (T/L) pathologies account for a significant portion of musculoskeletal injuries and disorders. Tissue engineering has emerged as a promising solution in the regeneration of both tissues. Specifically, the use of multipotent human mesenchymal stromal cells (hMSC) has shown great promise to serve as both a suitable cell source for tenogenic regeneration and a source of trophic factors to induce tenogenesis. Using four donor sets, we investigated the bidirectional paracrine tenogenic response between human hamstring tenocytes (hHT) and bone marrow-derived hMSC. Cell metabolic assays showed that only one hHT donor experienced sustained notable increases in cell metabolic activity during co-culture. Histological staining confirmed that co-culture induced elevated collagen protein levels in both cell types at varying time-points in two of four donor sets assessed. Gene expression analysis using qPCR showed the varied up-regulation of anabolic and catabolic markers involved in extracellular matrix maintenance for hMSC and hHT. Furthermore, analysis of hMSC/hHT co-culture secretome using a reporter cell line for TGF-β, a potent inducer of tenogenesis, revealed a trend of higher TGF-β bioactivity in hMSC secretome compared to hHT. Finally, hHT cytoskeletal immunostaining confirmed that both cell types released soluble factors capable of inducing favorable tenogenic morphology, comparable to control levels of soluble TGF-β1. These results suggest a potential for TGF-β-mediated signaling mechanism that is involved during the paracrine interplay between the two cell types that is reminiscent of T/L matrix remodeling/turnover. These findings have significant implications in the clinical use of hMSC for common T/L pathologies.
Collapse
Affiliation(s)
- Emmanuel C Ekwueme
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey.,MIRA Institute for Biomedical Technology and Technical Medicine, Department of Tissue Regeneration, University of Twente, Enschede, The Netherlands
| | - Jay V Shah
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Mahir Mohiuddin
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Corina A Ghebes
- MIRA Institute for Biomedical Technology and Technical Medicine, Department of Tissue Regeneration, University of Twente, Enschede, The Netherlands
| | - João F Crispim
- MIRA Institute for Biomedical Technology and Technical Medicine, Department of Tissue Regeneration, University of Twente, Enschede, The Netherlands
| | - Daniël B F Saris
- MIRA Institute for Biomedical Technology and Technical Medicine, Department of Tissue Regeneration, University of Twente, Enschede, The Netherlands.,Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hugo A M Fernandes
- MIRA Institute for Biomedical Technology and Technical Medicine, Department of Tissue Regeneration, University of Twente, Enschede, The Netherlands.,CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,UC-Biotech-Cantanhede, Cantanhede, Portugal
| | - Joseph W Freeman
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
134
|
Budi EH, Muthusamy BP, Derynck R. The insulin response integrates increased TGF-β signaling through Akt-induced enhancement of cell surface delivery of TGF-β receptors. Sci Signal 2015; 8:ra96. [PMID: 26420907 DOI: 10.1126/scisignal.aaa9432] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increased activity of transforming growth factor-β (TGF-β), which binds to and stimulates cell surface receptors, contributes to cancer progression and fibrosis by driving epithelial cells toward a migratory mesenchymal phenotype and increasing the abundance of extracellular matrix proteins. The abundance of TGF-β receptors at the cell surface determines cellular responsiveness to TGF-β, which is often produced by the same cells that have the receptors, and thus serves as an autocrine signal. We found that Akt-mediated phosphorylation of AS160, a RabGAP [guanosine triphosphatase (GTPase)-activating protein], promoted the translocation of TGF-β receptors from intracellular stores to the plasma membrane of mouse embryonic fibroblasts and NMuMG epithelial cells. Consequently, insulin, which is commonly used to treat hyperglycemia and activates Akt signaling, increased the amount of TGF-β receptors at the cell surface, thereby enhancing TGF-β responsiveness. This insulin-induced increase in autocrine TGF-β signaling contributed to insulin-induced gene expression responses, attenuated the epithelial phenotype, and promoted the migration of NMuMG cells. Furthermore, the enhanced delivery of TGF-β receptors at the cell surface enabled insulin to increase TGF-β-induced gene responses. The enhancement of TGF-β responsiveness in response to Akt activation may help to explain the biological effects of insulin, the progression of cancers in which Akt is activated, and the increased incidence of fibroses in diabetes.
Collapse
Affiliation(s)
- Erine H Budi
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143-0669, USA
| | - Baby-Periyanayaki Muthusamy
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143-0669, USA
| | - Rik Derynck
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143-0669, USA.
| |
Collapse
|
135
|
Stanley FM, Linder KM, Cardozo TJ. Statins Increase Plasminogen Activator Inhibitor Type 1 Gene Transcription through a Pregnane X Receptor Regulated Element. PLoS One 2015; 10:e0138097. [PMID: 26379245 PMCID: PMC4574702 DOI: 10.1371/journal.pone.0138097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/25/2015] [Indexed: 12/15/2022] Open
Abstract
Plasminogen activator inhibitor type 1 (PAI-1) is a multifunctional protein that has important roles in inflammation and wound healing. Its aberrant regulation may contribute to many disease processes such as heart disease. The PAI-1 promoter is responsive to multiple inputs including cytokines, growth factors, steroids and oxidative stress. The statin drugs, atorvastatin, mevastatin and rosuvastatin, increased basal and stimulated expression of the PAI-1 promoter 3-fold. A statin-responsive, nuclear hormone response element was previously identified in the PAI-1 promoter, but it was incompletely characterized. We characterized this direct repeat (DR) of AGGTCA with a 3-nucleotide spacer at -269/-255 using deletion and directed mutagenesis. Deletion or mutation of this element increased basal transcription from the promoter suggesting that it repressed PAI-1 transcription in the unliganded state. The half-site spacing and the ligand specificity suggested that this might be a pregnane X receptor (PXR) responsive element. Computational molecular docking showed that atorvastatin, mevastatin and rosuvastatin were structurally compatible with the PXR ligand-binding pocket in its agonist conformation. Experiments with Gal4 DNA binding domain fusion proteins showed that Gal4-PXR was activated by statins while other DR + 3 binding nuclear receptor fusions were not. Overexpression of PXR further enhanced PAI-1 transcription in response to statins. Finally, ChIP experiments using Halo-tagged PXR and RXR demonstrated that both components of the PXR-RXR heterodimer bound to this region of the PAI-1 promoter.
Collapse
Affiliation(s)
- Frederick M. Stanley
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York, United States of America
| | - Kathryn M. Linder
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
| | - Timothy J. Cardozo
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York, United States of America
| |
Collapse
|
136
|
Mann EH, Chambers ES, Chen YH, Richards DF, Hawrylowicz CM. 1α,25-dihydroxyvitamin D3 acts via transforming growth factor-β to up-regulate expression of immunosuppressive CD73 on human CD4+ Foxp3- T cells. Immunology 2015; 146:423-31. [PMID: 26251265 PMCID: PMC4610631 DOI: 10.1111/imm.12519] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/29/2015] [Accepted: 07/31/2015] [Indexed: 02/06/2023] Open
Abstract
Vitamin D deficiency is associated with increased incidence and severity of various immune-mediated diseases. Active vitamin D (1α,25-dihydroxyvitamin D3; 1,25(OH)2 D3) up-regulates CD4(+) T-cell expression of the purine ectonucleotidase CD39, a molecule that is associated with the generation of anti-inflammatory adenosine. Here we aimed to investigate the direct impact of 1,25(OH)2 D3 on expression of the downstream ecto-5'-nucleotidase CD73 by human CD4 T cells, and components of the transforming growth factor-β (TGF-β) pathway, which have been implicated in the modulation of CD73 by murine T cells. At 10(-8) to 10(-7) m, 1,25(OH)2 D3 significantly increased expression of CD73 on peripheral human CD4(+) T cells. Although 1,25(OH)2 D3 did not affect the mRNA expression of latent TGF-β1 , 1,25(OH)2 D3 did up-regulate expression of TGF-β-associated molecules [latency-associated peptide (LAP), glycophorin A repetitions predominant (GARP), GP96, neuropilin-1, thrombospondin-1 and αv integrin] which is likely to have contributed to the observed enhancement in TGF-β bioactivity. CD73 was highly co-expressed with LAP and GARP following 1,25(OH)2 D3 treatment, but unexpectedly, each of these cell surface molecules was expressed primarily on CD4(+) Foxp3(-) T cells, rather than CD4(+) Foxp3(+) T cells. Notably, neutralization of TGF-β significantly impaired 1,25(OH)2 D3-mediated induction of CD73. Collectively, we show that 1,25(OH)2 D3 enhances expression of CD73 on CD4(+) Foxp3(-) T cells in a process that is at least partially TGF-β-dependent. These data reveal an additional contributing mechanism by which vitamin D may be protective in immune-mediated disease.
Collapse
Affiliation(s)
- Elizabeth H Mann
- MRC and Asthma-UK Centre for Allergic Mechanisms in Asthma, King's College London, London, UK
| | - Emma S Chambers
- MRC and Asthma-UK Centre for Allergic Mechanisms in Asthma, King's College London, London, UK
| | - Yin-Huai Chen
- MRC and Asthma-UK Centre for Allergic Mechanisms in Asthma, King's College London, London, UK
| | - David F Richards
- MRC and Asthma-UK Centre for Allergic Mechanisms in Asthma, King's College London, London, UK
| | - Catherine M Hawrylowicz
- MRC and Asthma-UK Centre for Allergic Mechanisms in Asthma, King's College London, London, UK
| |
Collapse
|
137
|
Scharnweber D, Hübner L, Rother S, Hempel U, Anderegg U, Samsonov SA, Pisabarro MT, Hofbauer L, Schnabelrauch M, Franz S, Simon J, Hintze V. Glycosaminoglycan derivatives: promising candidates for the design of functional biomaterials. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2015; 26:232. [PMID: 26358319 DOI: 10.1007/s10856-015-5563-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/27/2015] [Indexed: 06/05/2023]
Abstract
Numerous biological processes (tissue formation, remodelling and healing) are strongly influenced by the cellular microenvironment. Glycosaminoglycans (GAGs) are important components of the native extracellular matrix (ECM) able to interact with biological mediator proteins. They can be chemically functionalized and thereby modified in their interaction profiles. Thus, they are promising candidates for functional biomaterials to control healing processes in particular in health-compromised patients. Biophysical studies show that the interaction profiles between mediator proteins and GAGs are strongly influenced by (i) sulphation degree, (ii) sulphation pattern, and (iii) composition and structure of the carbohydrate backbone. Hyaluronan derivatives demonstrate a higher binding strength in their interaction with biological mediators than chondroitin sulphate for a comparable sulphation degree. Furthermore sulphated GAG derivatives alter the interaction profile of mediator proteins with their cell receptors or solute native interaction partners. These results are in line with biological effects on cells relevant for wound healing processes. This is valid for solute GAGs as well as those incorporated in collagen-based artificial ECM (aECMs). Prominent effects are (i) anti-inflammatory, immunomodulatory properties towards macrophages/dendritic cells, (ii) enhanced osteogenic differentiation of human mesenchymal stromal cells, (iii) altered differentiation of fibroblasts to myofibroblasts, (iv) reduced osteoclast activity and (v) improved osseointegration of dental implants in minipigs. The findings of our consortium Transregio 67 contribute to an improved understanding of structure-function relationships of GAG derivatives in their interaction with mediator proteins and cells. This will enable the design of bioinspired, functional biomaterials to selectively control and promote bone and skin regeneration.
Collapse
Affiliation(s)
- Dieter Scharnweber
- Max Bergmann Center of Biomaterials, Institute of Materials Science, TU Dresden, Dresden, Germany.
| | - Linda Hübner
- Max Bergmann Center of Biomaterials, Institute of Materials Science, TU Dresden, Dresden, Germany
| | - Sandra Rother
- Max Bergmann Center of Biomaterials, Institute of Materials Science, TU Dresden, Dresden, Germany
| | - Ute Hempel
- Medical Department, Institute of Physiological Chemistry, TU Dresden, Dresden, Germany
| | - Ulf Anderegg
- Department of Dermatology, Venerology and Allergology, Leipzig University, Leipzig, Germany
| | | | | | - Lorenz Hofbauer
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine III, TU Dresden Medical Center, Dresden, Germany
| | | | - Sandra Franz
- Department of Dermatology, Venerology and Allergology, Leipzig University, Leipzig, Germany
| | - Jan Simon
- Department of Dermatology, Venerology and Allergology, Leipzig University, Leipzig, Germany
| | - Vera Hintze
- Max Bergmann Center of Biomaterials, Institute of Materials Science, TU Dresden, Dresden, Germany
| |
Collapse
|
138
|
O’Flaherty BM, Matar CG, Wakeman BS, Garcia A, Wilke CA, Courtney CL, Moore BB, Speck SH. CD8+ T Cell Response to Gammaherpesvirus Infection Mediates Inflammation and Fibrosis in Interferon Gamma Receptor-Deficient Mice. PLoS One 2015; 10:e0135719. [PMID: 26317335 PMCID: PMC4552722 DOI: 10.1371/journal.pone.0135719] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 07/24/2015] [Indexed: 02/05/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF), one of the most severe interstitial lung diseases, is a progressive fibrotic disorder of unknown etiology. However, there is growing appreciation for the role of viral infection in disease induction and/or progression. A small animal model of multi-organ fibrosis, which involves murine gammaherpesvirus (MHV68) infection of interferon gamma receptor deficient (IFNγR-/-) mice, has been utilized to model the association of gammaherpesvirus infections and lung fibrosis. Notably, several MHV68 mutants which fail to induce fibrosis have been identified. Our current study aimed to better define the role of the unique MHV68 gene, M1, in development of pulmonary fibrosis. We have previously shown that the M1 gene encodes a secreted protein which possesses superantigen-like function to drive the expansion and activation of Vβ4+ CD8+ T cells. Here we show that M1-dependent fibrosis is correlated with heightened levels of inflammation in the lung. We observe an M1-dependent cellular infiltrate of innate immune cells with most striking differences at 28 days-post infection. Furthermore, in the absence of M1 protein expression we observed reduced CD8+ T cells and MHV68 epitope specific CD8+ T cells to the lungs-despite equivalent levels of viral replication between M1 null and wild type MHV68. Notably, backcrossing the IFNγR-/- onto the Balb/c background, which has previously been shown to exhibit weak MHV68-driven Vβ4+ CD8+ T cell expansion, eliminated MHV68-induced fibrosis-further implicating the activated Vβ4+ CD8+ T cell population in the induction of fibrosis. We further addressed the role that CD8+ T cells play in the induction of fibrosis by depleting CD8+ T cells, which protected the mice from fibrotic disease. Taken together these findings are consistent with the hypothesized role of Vβ4+ CD8+ T cells as mediators of fibrotic disease in IFNγR-/- mice.
Collapse
Affiliation(s)
- Brigid M. O’Flaherty
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States of America
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Caline G. Matar
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States of America
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Brian S. Wakeman
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States of America
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States of America
| | - AnaPatricia Garcia
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta GA, United States of America
| | - Carol A. Wilke
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Cynthia L. Courtney
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta GA, United States of America
| | - Bethany B. Moore
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Samuel H. Speck
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States of America
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States of America
| |
Collapse
|
139
|
Schmidt M, Gutknecht D, Simon JC, Schulz JN, Eckes B, Anderegg U, Saalbach A. Controlling the Balance of Fibroblast Proliferation and Differentiation: Impact of Thy-1. J Invest Dermatol 2015; 135:1893-1902. [DOI: 10.1038/jid.2015.86] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/03/2015] [Accepted: 02/19/2015] [Indexed: 11/09/2022]
|
140
|
Jenkins MH, Croteau W, Mullins DW, Brinckerhoff CE. The BRAF(V600E) inhibitor, PLX4032, increases type I collagen synthesis in melanoma cells. Matrix Biol 2015; 48:66-77. [PMID: 25989506 PMCID: PMC5048745 DOI: 10.1016/j.matbio.2015.05.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 01/08/2023]
Abstract
Vertical growth phase (VGP) melanoma is frequently metastatic, a process mediated by changes in gene expression, which are directed by signal transduction pathways in the tumor cells. A prominent signaling pathway is the Ras-Raf-Mek-Erk MAPK pathway, which increases expression of genes that promote melanoma progression. Many melanomas harbor a mutation in this pathway, BRAF(V600E), which constitutively activates MAPK signaling and expression of downstream target genes that facilitate tumor progression. In BRAF(V600E) melanoma, the small molecule inhibitor, vemurafenib (PLX4032), has revolutionized therapy for melanoma by inducing rapid tumor regression. This compound down-regulates the expression of many genes. However, in this study, we document that blocking the Ras-Raf-Mek-Erk MAPK pathway, either with an ERK (PLX4032) or a MEK (U1026) signaling inhibitor, in BRAF(V600E) human and murine melanoma cell lines increases collagen synthesis in vitro and collagen deposition in vivo. Since TGFß signaling is a major mediator of collagen synthesis, we examined whether blocking TGFß signaling with a small molecule inhibitor would block this increase in collagen. However, there was minimal reduction in collagen synthesis in response to blocking TGFß signaling, suggesting additional mechanism(s), which may include activation of the p38 MAPK pathway. Presently, it is unclear whether this increased collagen synthesis and deposition in melanomas represent a therapeutic benefit or an unwanted "off target" effect of inhibiting the Ras-Raf-Erk-Mek pathway.
Collapse
Affiliation(s)
- Molly H Jenkins
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States; Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States.
| | - Walburga Croteau
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States
| | - David W Mullins
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States
| | - Constance E Brinckerhoff
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States; Department of Biochemistry, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States
| |
Collapse
|
141
|
Worthington JJ, Kelly A, Smedley C, Bauché D, Campbell S, Marie JC, Travis MA. Integrin αvβ8-Mediated TGF-β Activation by Effector Regulatory T Cells Is Essential for Suppression of T-Cell-Mediated Inflammation. Immunity 2015; 42:903-15. [PMID: 25979421 PMCID: PMC4448149 DOI: 10.1016/j.immuni.2015.04.012] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 12/22/2014] [Accepted: 04/23/2015] [Indexed: 01/19/2023]
Abstract
Regulatory T (Treg) cells play a pivotal role in suppressing self-harmful T cell responses, but how Treg cells mediate suppression to maintain immune homeostasis and limit responses during inflammation is unclear. Here we show that effector Treg cells express high amounts of the integrin αvβ8, which enables them to activate latent transforming growth factor-β (TGF-β). Treg-cell-specific deletion of integrin αvβ8 did not result in a spontaneous inflammatory phenotype, suggesting that this pathway is not important in Treg-cell-mediated maintenance of immune homeostasis. However, Treg cells lacking expression of integrin αvβ8 were unable to suppress pathogenic T cell responses during active inflammation. Thus, our results identify a mechanism by which Treg cells suppress exuberant immune responses, highlighting a key role for effector Treg-cell-mediated activation of latent TGF-β in suppression of self-harmful T cell responses during active inflammation. Human and mouse effector Treg cells express functional TGF-β-activating integrin αvβ8 Treg cell integrin αvβ8-mediated TGF-β activation is not needed for T cell homeostasis Integrin αvβ8 expression by Treg cells suppresses active inflammation Pathway could be targeted to promote Treg-cell-mediated suppression of inflammation
Collapse
Affiliation(s)
- John J Worthington
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester M13 9NT, UK; Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK; Manchester Immunology Group, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK.
| | - Aoife Kelly
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester M13 9NT, UK; Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK; Manchester Immunology Group, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Catherine Smedley
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester M13 9NT, UK; Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK; Manchester Immunology Group, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - David Bauché
- Immunology Virology and Inflammation Department, CRCL, UMR INSERM1052, CNRS 5286, Centre Léon Bérard, 28 rue Laennec, 69373 Cedex 08 Lyon, France; Université Lyon 1, 69000 Lyon, France; Labex DEVweCAN, 69008 Lyon, France; TGFβ and Immuno-evasion Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Simon Campbell
- Gastroenterology Unit, Manchester Royal Infirmary, Central Manchester University Hospital NHS Foundation Trust, Manchester M13 9WL, UK
| | - Julien C Marie
- Immunology Virology and Inflammation Department, CRCL, UMR INSERM1052, CNRS 5286, Centre Léon Bérard, 28 rue Laennec, 69373 Cedex 08 Lyon, France; Université Lyon 1, 69000 Lyon, France; Labex DEVweCAN, 69008 Lyon, France; TGFβ and Immuno-evasion Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Mark A Travis
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester M13 9NT, UK; Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK; Manchester Immunology Group, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK.
| |
Collapse
|
142
|
Taniguchi T, Asano Y, Akamata K, Noda S, Takahashi T, Ichimura Y, Toyama T, Trojanowska M, Sato S. Fibrosis, vascular activation, and immune abnormalities resembling systemic sclerosis in bleomycin-treated Fli-1-haploinsufficient mice. Arthritis Rheumatol 2015; 67:517-26. [PMID: 25385187 DOI: 10.1002/art.38948] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 11/04/2014] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Fli-1, a potential predisposing factor for systemic sclerosis (SSc), is constitutively down-regulated in the lesional skin of patients with SSc by an epigenetic mechanism. To investigate the impact of Fli-1 deficiency on the induction of an SSc phenotype in various cell types, we generated bleomycin-induced skin fibrosis in Fli-1(+/-) mice and investigated the molecular mechanisms underlying its phenotypic alterations. METHODS Messenger RNA (mRNA) levels and protein expression of target molecules were examined by quantitative reverse transcription-polymerase chain reaction and immunostaining. Transforming growth factor β (TGFβ) bioassay was used to evaluate the activation of latent TGFβ. The binding of Fli-1 to the target gene promoters was assessed with chromatin immunoprecipitation. RESULTS Bleomycin induced more severe dermal fibrosis in Fli-1(+/-) mice than in wild-type mice. Fli-1 haploinsufficiency activated dermal fibroblasts via the up-regulation of αvβ3 and αvβ5 integrins and activation of latent TGFβ. Dermal fibrosis in Fli-1(+/-) mice was also attributable to endothelial-to-mesenchymal transition, which is directly induced by Fli-1 deficiency and amplified by bleomycin. Th2/Th17-skewed inflammation and increased infiltration of mast cells and macrophages were seen, partly due to the altered expression of cell adhesion molecules in endothelial cells as well as the induction of the skin chemokines. Fli-1(+/-) mouse macrophages preferentially differentiated into an M2 phenotype upon stimulation with interleukin-4 (IL-4) or IL-13. CONCLUSION Our findings provide strong evidence for the fundamental role of Fli-1 deficiency in inducing SSc-like phenotypic alterations in dermal fibroblasts, endothelial cells, and macrophages in a manner consistent with human disease.
Collapse
|
143
|
Su CT, Huang JW, Chiang CK, Lawrence EC, Levine KL, Dabovic B, Jung C, Davis EC, Madan-Khetarpal S, Urban Z. Latent transforming growth factor binding protein 4 regulates transforming growth factor beta receptor stability. Hum Mol Genet 2015; 24:4024-36. [PMID: 25882708 DOI: 10.1093/hmg/ddv139] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 04/13/2015] [Indexed: 01/17/2023] Open
Abstract
Mutations in the gene for the latent transforming growth factor beta binding protein 4 (LTBP4) cause autosomal recessive cutis laxa type 1C. To understand the molecular disease mechanisms of this disease, we investigated the impact of LTBP4 loss on transforming growth factor beta (TGFβ) signaling. Despite elevated extracellular TGFβ activity, downstream signaling molecules of the TGFβ pathway, including pSMAD2 and pERK, were down-regulated in LTBP4 mutant human dermal fibroblasts. In addition, TGFβ receptors 1 and 2 (TGFBR1 and TGFBR2) were reduced at the protein but not at the ribonucleic acid level. Treatment with exogenous TGFβ1 led to an initially rapid increase in SMAD2 phosphorylation followed by a sustained depression of phosphorylation and receptor abundance. In mutant cells TGFBR1 was co-localized with lysosomes. Treatment with a TGFBR1 kinase inhibitor, endocytosis inhibitors or a lysosome inhibitor, normalized the levels of TGFBR1 and TGFBR2. Co-immunoprecipitation demonstrated a molecular interaction between LTBP4 and TGFBR2. Knockdown of LTBP4 reduced TGFβ receptor abundance and signaling in normal cells and supplementation of recombinant LTBP4 enhanced these measures in mutant cells. In a mouse model of Ltbp4 deficiency, reduced TGFβ signaling and receptor levels were normalized upon TGFBR1 kinase inhibitor treatment. Our results show that LTBP4 interacts with TGFBR2 and stabilizes TGFβ receptors by preventing their endocytosis and lysosomal degradation in a ligand-dependent and receptor kinase activity-dependent manner. These findings identify LTBP4 as a key molecule required for the stability of the TGFβ receptor complex, and a new mechanism by which the extracellular matrix regulates cytokine receptor signaling.
Collapse
Affiliation(s)
- Chi-Ting Su
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Crabtree Hall A300, Pittsburgh, PA 15261, USA
| | - Jenq-Wen Huang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Medical College and Hospital, Taipei 100, Taiwan
| | - Chih-Kang Chiang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Medical College and Hospital, Taipei 100, Taiwan
| | - Elizabeth C Lawrence
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Crabtree Hall A300, Pittsburgh, PA 15261, USA
| | - Kara L Levine
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Crabtree Hall A300, Pittsburgh, PA 15261, USA
| | - Branka Dabovic
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Christine Jung
- Practice of Human Genetics, Karlsruhe 76133, Germany, Synlab MVZ Human Genetics, Mannheim 68163, Germany
| | - Elaine C Davis
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 2B2, Canada and
| | - Suneeta Madan-Khetarpal
- Division of Medical Genetics, Children's Hospital of Pittsburgh of UMPC, Pittsburgh, PA 15224, USA
| | - Zsolt Urban
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Crabtree Hall A300, Pittsburgh, PA 15261, USA,
| |
Collapse
|
144
|
Pellicciotta I, Marciscano AE, Hardee ME, Francis D, Formenti S, Barcellos-Hoff MH. Development of a novel multiplexed assay for quantification of transforming growth factor-β (TGF-β). Growth Factors 2015; 33:79-91. [PMID: 25586866 DOI: 10.3109/08977194.2014.999367] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Changes in activity or levels of transforming growth factor-β (TGF-β) are associated with a variety of diseases; however, measurement of TGF-β in biological fluids is highly variable. TGF-β is biologically inert when associated with its latency-associated peptide (LAP). Most available immunoassays require exogenous activation by acid/heat to release TGF-β from the latent complex. We developed a novel electrochemiluminescence-based multiplexed assay on the MesoScale Discovery® platform that eliminates artificial activation, simultaneously measures both active TGF-β1 and LAP1 and includes an internal control for platelet-derived TGF-β contamination in blood specimens. We optimized this assay to evaluate plasma levels as a function of activation type and clinical specimen preparation. We determined that breast cancer patients' plasma have higher levels of circulating latent TGF-β (LTGF-β) as measured by LAP1 than healthy volunteers (p < 0.0001). This assay provides a robust tool for correlative studies of LTGF-β levels with disease, treatment outcomes and toxicity with a broad clinical applicability.
Collapse
|
145
|
Dayer C, Stamenkovic I. Recruitment of Matrix Metalloproteinase-9 (MMP-9) to the Fibroblast Cell Surface by Lysyl Hydroxylase 3 (LH3) Triggers Transforming Growth Factor-β (TGF-β) Activation and Fibroblast Differentiation. J Biol Chem 2015; 290:13763-78. [PMID: 25825495 DOI: 10.1074/jbc.m114.622274] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Indexed: 12/27/2022] Open
Abstract
Solid tumor growth triggers a wound healing response. Similar to wound healing, fibroblasts in the tumor stroma differentiate into myofibroblasts (also referred to as cancer-associated fibroblasts) primarily, but not exclusively, in response to transforming growth factor-β (TGF-β). Myofibroblasts in turn enhance tumor progression by remodeling the stroma. Among proteases implicated in stroma remodeling, matrix metalloproteinases (MMPs), including MMP-9, play a prominent role. Recent evidence indicates that MMP-9 recruitment to the tumor cell surface enhances tumor growth and invasion. In the present work, we addressed the potential relevance of MMP-9 recruitment to and activity at the surface of fibroblasts. We show that recruitment of MMP-9 to the fibroblast cell surface occurs through its fibronectin-like (FN) domain and that the molecule responsible for the recruitment is lysyl hydroxylase 3 (LH3). Functional assays suggest that both pro- and active MMP-9 trigger α-smooth muscle actin expression in cultured fibroblasts, reflecting myofibroblast differentiation, possibly as a result of TGF-β activation. Moreover, the recombinant FN domain inhibited both MMP-9-induced TGF-β activation and α-smooth muscle actin expression by displacing MMP-9 from the fibroblast cell surface. Together our results uncover LH3 as a new docking receptor of MMP-9 on the fibroblast cell surface and demonstrate that the MMP-9 FN domain is essential for the interaction. They also show that the recombinant FN domain inhibits MMP-9-induced TGF-β activation and fibroblast differentiation, providing a potentially attractive therapeutic reagent toward attenuating tumor progression where MMP-9 activity is strongly implicated.
Collapse
Affiliation(s)
- Cynthia Dayer
- From the Division of Experimental Pathology, Institute of Pathology, CHUV, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 25, Lausanne CH1011, Switzerland
| | - Ivan Stamenkovic
- From the Division of Experimental Pathology, Institute of Pathology, CHUV, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 25, Lausanne CH1011, Switzerland
| |
Collapse
|
146
|
Bultmann-Mellin I, Conradi A, Maul AC, Dinger K, Wempe F, Wohl AP, Imhof T, Wunderlich FT, Bunck AC, Nakamura T, Koli K, Bloch W, Ghanem A, Heinz A, von Melchner H, Sengle G, Sterner-Kock A. Modeling autosomal recessive cutis laxa type 1C in mice reveals distinct functions for Ltbp-4 isoforms. Dis Model Mech 2015; 8:403-15. [PMID: 25713297 PMCID: PMC4381339 DOI: 10.1242/dmm.018960] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/16/2015] [Indexed: 01/03/2023] Open
Abstract
Recent studies have revealed an important role for LTBP-4 in elastogenesis. Its mutational inactivation in humans causes autosomal recessive cutis laxa type 1C (ARCL1C), which is a severe disorder caused by defects of the elastic fiber network. Although the human gene involved in ARCL1C has been discovered based on similar elastic fiber abnormalities exhibited by mice lacking the short Ltbp-4 isoform (Ltbp4S(-/-)), the murine phenotype does not replicate ARCL1C. We therefore inactivated both Ltbp-4 isoforms in the mouse germline to model ARCL1C. Comparative analysis of Ltbp4S(-/-) and Ltbp4-null (Ltbp4(-/-)) mice identified Ltbp-4L as an important factor for elastogenesis and postnatal survival, and showed that it has distinct tissue expression patterns and specific molecular functions. We identified fibulin-4 as a previously unknown interaction partner of both Ltbp-4 isoforms and demonstrated that at least Ltbp-4L expression is essential for incorporation of fibulin-4 into the extracellular matrix (ECM). Overall, our results contribute to the current understanding of elastogenesis and provide an animal model of ARCL1C.
Collapse
Affiliation(s)
- Insa Bultmann-Mellin
- Center for Experimental Medicine, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Anne Conradi
- Center for Experimental Medicine, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Alexandra C Maul
- Center for Experimental Medicine, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Katharina Dinger
- Center for Experimental Medicine, Medical Faculty, University of Cologne, 50931 Cologne, Germany. Department of Pediatrics and Adolescent Medicine, Medical Faculty, University of Cologne, 50937 Cologne, Germany
| | - Frank Wempe
- Department of Molecular Hematology, University of Frankfurt Medical School, 60590 Frankfurt am Main, Germany
| | - Alexander P Wohl
- Center for Biochemistry, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Thomas Imhof
- Center for Biochemistry, Medical Faculty, University of Cologne, 50931 Cologne, Germany. Institute for Dental Research and Oral Musculoskeletal Biology, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - F Thomas Wunderlich
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany. Max Planck Institute for Metabolism Research, 50931 Cologne, Germany. Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Alexander C Bunck
- Department of Radiology, Medical Faculty, University of Cologne, 50937 Cologne, Germany
| | - Tomoyuki Nakamura
- Department of Pharmacology, Kansai Medical University, Osaka 570-8506, Japan
| | - Katri Koli
- Research Programs Unit and Transplantation Laboratory, Haartman Institute, University of Helsinki, 00014 Helsinki, Finland
| | - Wilhelm Bloch
- Institute of Cardiology and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany
| | - Alexander Ghanem
- Department of Medicine/Cardiology, University of Bonn, 53127 Bonn, Germany
| | - Andrea Heinz
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Harald von Melchner
- Department of Molecular Hematology, University of Frankfurt Medical School, 60590 Frankfurt am Main, Germany
| | - Gerhard Sengle
- Center for Biochemistry, Medical Faculty, University of Cologne, 50931 Cologne, Germany. Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Anja Sterner-Kock
- Center for Experimental Medicine, Medical Faculty, University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
147
|
HGF Modulates Actin Cytoskeleton Remodeling and Contraction in Testicular Myoid Cells. Biomedicines 2015; 3:89-109. [PMID: 28536401 PMCID: PMC5344232 DOI: 10.3390/biomedicines3010089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/10/2014] [Accepted: 01/21/2015] [Indexed: 12/05/2022] Open
Abstract
The presence of the HGF/Met system in the testicular myoid cells was first discovered by our group. However, the physiological role of this pathway remains poorly understood. We previously reported that HGF increases uPA secretion and TGF-β activation in cultured tubular fragments and that HGF is maximally expressed at Stages VII–VIII of the seminiferous epithelium cycle, when myoid cell contraction occurs. It is well known that the HGF/Met pathway is involved in cytoskeletal remodeling; moreover, the interaction of uPA with its receptor, uPAR, as well as the activation of TGF-β have been reported to be related to the actin cytoskeleton contractility of smooth muscle cells. Herein, we report that HGF induces actin cytoskeleton remodeling in vitro in isolated myoid cells and myoid cell contraction in cultured seminiferous tubules. To better understand these phenomena, we evaluated: (1) the regulation of the uPA machinery in isolated myoid cells after HGF administration; and (2) the effect of uPA or Met inhibition on HGF-treated tubular fragments. Because uPA activates latent TGF-β, the secretion of this factor was also evaluated. We found that both uPA and TGF-β activation increase after HGF administration. In testicular tubular fragments, HGF-induced TGF-β activation and myoid cell contraction are abrogated by uPA or Met inhibitor administration.
Collapse
|
148
|
Rana MK, Srivastava J, Yang M, Chen CS, Barber DL. Hypoxia increases the abundance but not the assembly of extracellular fibronectin during epithelial cell transdifferentiation. J Cell Sci 2015; 128:1083-9. [PMID: 25616899 DOI: 10.1242/jcs.155036] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Increased production and assembly of extracellular matrix proteins during transdifferentiation of epithelial cells to a mesenchymal phenotype contributes to diseases such as renal and pulmonary fibrosis. TGF-β and hypoxia, two cues that initiate injury-induced fibrosis, caused human kidney cells to develop a mesenchymal phenotype, including increased fibronectin expression and secretion. However, upon hypoxia, assembled extracellular fibronectin fibrils were mostly absent, whereas treatment with TGF-β led to abundant fibrils. Fibrillogenesis required cell-generated force and tension. TGF-β, but not hypoxia, increased cell contractility, as determined by phosphorylation of myosin light chain and quantifying force and tension generated by cells plated on engineered elastomeric microposts. Additionally, TGF-β, but not hypoxia, increased the activation of integrins. However, experimentally activating integrins markedly increased the levels of phosphorylated myosin light chain and fibronectin fibril assembly upon hypoxia. Our findings show that deficient integrin activation and subsequent lack of cell contractility are mechanisms that mediate a lack of fibrillogenesis upon hypoxia and they challenge current views on oxygen deprivation being sufficient for fibrosis.
Collapse
Affiliation(s)
- Manish K Rana
- Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA Department of Biomedical Engineering, Boston University and Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Jyoti Srivastava
- Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA
| | - Michael Yang
- Department of Biomedical Engineering, Boston University and Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University and Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Diane L Barber
- Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
149
|
Sapudom J, Rubner S, Martin S, Thoenes S, Anderegg U, Pompe T. The interplay of fibronectin functionalization and TGF-β1 presence on fibroblast proliferation, differentiation and migration in 3D matrices. Biomater Sci 2015; 3:1291-301. [DOI: 10.1039/c5bm00140d] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
TGF-β1 dependent fibroblast behaviour in a wound healing context is mimicked by topologically and mechanically defined collagen matrices with fibronectin functionalization.
Collapse
Affiliation(s)
- Jiranuwat Sapudom
- Institute of Biochemistry
- Faculty of Biosciences
- Pharmacy and Psychology
- Universität Leipzig
- Leipzig 04103
| | - Stefan Rubner
- Institute of Biochemistry
- Faculty of Biosciences
- Pharmacy and Psychology
- Universität Leipzig
- Leipzig 04103
| | - Steve Martin
- Institute of Biochemistry
- Faculty of Biosciences
- Pharmacy and Psychology
- Universität Leipzig
- Leipzig 04103
| | - Stephan Thoenes
- Department of Dermatology
- Venereology and Allergology
- Universitätsklinikum Leipzig
- Leipzig 04103
- Germany
| | - Ulf Anderegg
- Department of Dermatology
- Venereology and Allergology
- Universitätsklinikum Leipzig
- Leipzig 04103
- Germany
| | - Tilo Pompe
- Institute of Biochemistry
- Faculty of Biosciences
- Pharmacy and Psychology
- Universität Leipzig
- Leipzig 04103
| |
Collapse
|
150
|
Ballesteros A, Mentink-Kane MM, Warren J, Kaplan GG, Dveksler GS. Induction and activation of latent transforming growth factor-β1 are carried out by two distinct domains of pregnancy-specific glycoprotein 1 (PSG1). J Biol Chem 2014; 290:4422-31. [PMID: 25548275 DOI: 10.1074/jbc.m114.597518] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Pregnancy-specific glycoproteins (PSGs) are a family of Ig-like proteins secreted by specialized placental cells. The PSG1 structure is composed of a single Ig variable region-like N-terminal domain and three Ig constant region-like domains termed A1, A2, and B2. Members of the human and murine PSG family have been shown to induce anti-inflammatory cytokines from monocytes and macrophages and to stimulate angiogenesis. We recently showed that recombinant forms of PSG1 (PSG1-Fc and PSG1-His) and PSG1 purified from the serum of pregnant women are associated with the immunoregulatory cytokine TGF-β1 and activated latent TGF-β1. Here, we sought to examine the requirement of specific PSG1 domains in the activation of latent TGF-β1. Plasmon surface resonance studies showed that PSG1 directly bound to the small latent complex and to the latency-associated peptide of TGF-β1 and that this binding was mediated through the B2 domain. Furthermore, the B2 domain alone was sufficient for activating the small latent complex. In separate experiments, we found that the PSG1-mediated induction of TGF-β1 secretion in macrophages was dependent on the N-terminal domain. Mutagenesis analysis revealed that four amino acids (LYHY) of the CC' loop of the N-terminal domain were required for induction of latent TGF-β1 secretion. Together, our results show that two distinct domains of PSG1 are involved in the regulation of TGF-β1 and provide a mechanistic framework for how PSGs modulate the immunoregulatory environment at the maternal-fetal interface for successful pregnancy outcome.
Collapse
Affiliation(s)
- Angela Ballesteros
- From the Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland 20892 and
| | - Margaret M Mentink-Kane
- the Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
| | - James Warren
- the Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
| | - Gerardo G Kaplan
- From the Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland 20892 and
| | - Gabriela S Dveksler
- the Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
| |
Collapse
|