1
|
Silva TA, Thomas D, Siqueira-Neto JL, Calvet CM. Pirfenidone Prevents Heart Fibrosis during Chronic Chagas Disease Cardiomyopathy. Int J Mol Sci 2024; 25:7302. [PMID: 39000409 PMCID: PMC11242150 DOI: 10.3390/ijms25137302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/23/2023] [Accepted: 12/28/2023] [Indexed: 07/16/2024] Open
Abstract
Cardiac fibrosis is a severe outcome of Chagas disease (CD), caused by the protozoan Trypanosoma cruzi. Clinical evidence revealed a correlation between fibrosis levels with impaired cardiac performance in CD patients. Therefore, we sought to analyze the effect of inhibitors of TGF-β (pirfenidone), p38-MAPK (losmapimod) and c-Jun (SP600125) on the modulation of collagen deposition in cardiac fibroblasts (CF) and in vivo models of T. cruzi chronic infection. Sirius Red/Fast Green dye was used to quantify both collagen expression and total protein amount, assessing cytotoxicity. The compounds were also used to treat C57/Bl6 mice chronically infected with T. cruzi, Brazil strain. We identified an anti-fibrotic effect in vitro for pirfenidone (TGF-β inhibitor, IC50 114.3 μM), losmapimod (p38 inhibitor, IC50 17.6 μM) and SP600125 (c-Jun inhibitor, IC50 3.9 μM). This effect was independent of CF proliferation since these compounds do not affect T. cruzi-induced host cell multiplication as measured by BrdU incorporation. Assays of chronic infection of mice with T. cruzi have shown a reduction in heart collagen by pirfenidone. These results propose a novel approach to fibrosis therapy in CD, with the prospect of repurposing pirfenidone to prevent the onset of ECM accumulation in the hearts of the patients.
Collapse
Affiliation(s)
- Tatiana Araújo Silva
- Cellular Ultrastructure Laboratory, Oswaldo Cruz Institute (IOC), FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil;
| | - Diane Thomas
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA; (D.T.); (J.L.S.-N.)
| | - Jair L. Siqueira-Neto
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA; (D.T.); (J.L.S.-N.)
| | - Claudia Magalhaes Calvet
- Cellular Ultrastructure Laboratory, Oswaldo Cruz Institute (IOC), FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil;
| |
Collapse
|
2
|
Wijewardhane PR, Wells A, Muhoberac M, Leung KP, Chopra G. Modeling Molecular Mechanisms of Pirfenidone Interaction with Kinases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586235. [PMID: 38585747 PMCID: PMC10996454 DOI: 10.1101/2024.03.22.586235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Scar formation is a process that occurs due to increased collagen deposition and uncontrolled inflammation. Previous studies have demonstrated that Pirfenidone (Pf), an FDA approved anti-inflammatory and antifibrotic drug can reduce inflammation in vivo as well as regulate activation of LPS-stimulated neutrophils. However, the molecular level mechanism of Pf's action is not well understood. Here, we used neural networks to identify new targets and molecular modeling methods to investigate the Pf's action pathways at the molecular level that are related to its ability to reduce both the inflammatory and remodeling phases of the wound healing process. Out of all the potential targets identified, both molecular docking and molecular dynamics results suggest that Pf has a noteworthy binding preference towards the active conformation of the p38 mitogen activated protein kinase-14 (MAPK14) and it is potentially a type I inhibitor-like molecule. In addition to p38 MAPK (MAPK14), additional potential targets of Pf include AKT1, MAP3K4, MAP2K3, MAP2K6, MSK2, MAP2K2, ERK1, ERK2, and PDK1. We conclude that several proteins/kinases, rather than a single target, are involved in Pf's wound healing ability to regulate signaling, inflammation, and proliferation.
Collapse
Affiliation(s)
| | - Adrienne Wells
- Combat Wound Care Group, US Army Institute of Surgical Research, 3650 Chambers Pass, Bldg 3610, JBSA Fort Sam Houston, TX 78234, USA
| | - Matthew Muhoberac
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA
| | - Kai P. Leung
- Combat Wound Care Group, US Army Institute of Surgical Research, 3650 Chambers Pass, Bldg 3610, JBSA Fort Sam Houston, TX 78234, USA
| | - Gaurav Chopra
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907, USA
- Department of Computer Science (by courtesy), Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Drug Discovery, 720 Clinic Drive, West Lafayette, IN 47907, USA
- Purdue Institute for Cancer Research, Purdue Institute for Integrative Neuroscience, Purdue Institute of Inflammation, Immunology and Infectious Disease Purdue University, West Lafayette, IN 47907, USA
- Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
3
|
Meng W, Jiang Z, Wang J, Chen X, Chen B, Cai B, Zhou Y, Ma L, Guan Y. Inhibition of urethral stricture by a catheter loaded with nanoparticle/ pirfenidone complexes. Front Bioeng Biotechnol 2023; 11:1254621. [PMID: 37954024 PMCID: PMC10639154 DOI: 10.3389/fbioe.2023.1254621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/13/2023] [Indexed: 11/14/2023] Open
Abstract
Background: Urethral strictures are common injurious conditions of the urinary system. Reducing and preventing urethral strictures has become a hot and challenging topic for urological surgeons and related researchers. In this study, we developed a catheter loaded with nanoparticle/pirfenidone (NP/PFD) complexes and evaluated its effectiveness at inhibiting urethral stricture in rabbits, providing more references for the clinical prevention and reduction of urethral stenosis. Methods: Twelve adult male New Zealand rabbits were selected and divided into the following four groups in a ratio of 1:1:1:1 using the random number table method: Group A, sham; Group B, urethral stricture (US); Group C, US + unmodified catheter; and Group D, US + NP/PFD catheter. On the 30th day after modelling, retrograde urethrography was performed to evaluate urethral stricture formation, and histopathological examination was performed on the tissues of the corresponding surgical site. Meanwhile, changes in the expression level of Transforming growth factor β1 (TGF-β1) in the tissues were detected by immunohistochemistry. Results: The NP/PFD complexes adhered uniformly to the catheter surface. They remained on the surface of the catheter after insertion into the urethra. In addition, the NP/PFD complexes spread into the urethral epithelium 2 weeks after surgery. Ultimately, urethral strictures were significantly reduced with the placement of the NP/PFD complex catheter. Conclusion: Our catheter loaded with NP/PFD complexes effectively delivered PFD to the urethral epithelium through continuous local delivery, thereby reducing fibrosis and stricture after urethral injury, which may be associated with the inhibition of TGF-β1 expression.
Collapse
Affiliation(s)
- Wei Meng
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Zhaosheng Jiang
- Department of Urology, Nantong TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Nantong, China
| | - Jiahao Wang
- Department of Urology, Wuxi Hospital Affiliated to the Nanjing University of Chinese Medicine, Wuxi, China
| | - Xiaohua Chen
- Department of Imaging, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Bo Chen
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Bo Cai
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Youlang Zhou
- Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Limin Ma
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Yangbo Guan
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| |
Collapse
|
4
|
Tottoli EM, Benedetti L, Riva F, Chiesa E, Pisani S, Bruni G, Genta I, Conti B, Ceccarelli G, Dorati R. Electrospun Fibers Loaded with Pirfenidone: An Innovative Approach for Scar Modulation in Complex Wounds. Polymers (Basel) 2023; 15:4045. [PMID: 37896289 PMCID: PMC10610295 DOI: 10.3390/polym15204045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/03/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Hypertrophic scars (HTSs) are pathological structures resulting from chronic inflammation during the wound healing process, particularly in complex injuries like burns. The aim of this work is to propose Biofiber PF (biodegradable fiber loaded with Pirfenidone 1.5 w/w), an electrospun advanced dressing, as a solution for HTSs treatment in complex wounds. Biofiber has a 3-day antifibrotic action to modulate the fibrotic process and enhance physiological healing. Its electrospun structure consists of regular well-interconnected Poly-L-lactide-co-poly-ε-caprolactone (PLA-PCL) fibers (size 2.83 ± 0.46 µm) loaded with Pirfenidone (PF, 1.5% w/w), an antifibrotic agent. The textured matrix promotes the exudate balance through mild hydrophobic wettability behavior (109.3 ± 2.3°), and an appropriate equilibrium between the absorbency % (610.2 ± 171.54%) and the moisture vapor transmission rate (0.027 ± 0.036 g/min). Through its finer mechanical properties, Biofiber PF is conformable to the wound area, promoting movement and tissue oxygenation. These features also enhance the excellent elongation (>500%) and tenacity, both in dry and wet conditions. The ancillary antifibrotic action of PF on hypertrophic scar fibroblast (HSF) for 3 days downregulates the cell proliferation over time and modulates the gene expression of transforming growth factor β1 (TGF-β1) and α-smooth muscle actin (α-SMA) at 48-72 h. After 6 days of treatment, a decrement of α-SMA protein levels was detected, proving the potential of biofiber as a valid therapeutic treatment for HTSs in an established wound healing process.
Collapse
Affiliation(s)
- Erika Maria Tottoli
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (E.M.T.); (E.C.); (S.P.); (I.G.); (B.C.)
| | - Laura Benedetti
- Department of Public Health, Experimental Medicine and Forensic, Human Anatomy Unit, University of Pavia, 27100 Pavia, Italy; (L.B.); (G.C.)
- CHT Center for Health Technologies, University of Pavia, 27100 Pavia, Italy
| | - Federica Riva
- Department of Public Health, Experimental Medicine and Forensic, Histology and Embryology Unit, University of Pavia, 27100 Pavia, Italy;
| | - Enrica Chiesa
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (E.M.T.); (E.C.); (S.P.); (I.G.); (B.C.)
| | - Silvia Pisani
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (E.M.T.); (E.C.); (S.P.); (I.G.); (B.C.)
| | - Giovanna Bruni
- Physical-Chemistry Section, Department of Chemistry, University of Pavia, 27100 Pavia, Italy;
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (E.M.T.); (E.C.); (S.P.); (I.G.); (B.C.)
- CHT Center for Health Technologies, University of Pavia, 27100 Pavia, Italy
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (E.M.T.); (E.C.); (S.P.); (I.G.); (B.C.)
- CHT Center for Health Technologies, University of Pavia, 27100 Pavia, Italy
| | - Gabriele Ceccarelli
- Department of Public Health, Experimental Medicine and Forensic, Human Anatomy Unit, University of Pavia, 27100 Pavia, Italy; (L.B.); (G.C.)
- CHT Center for Health Technologies, University of Pavia, 27100 Pavia, Italy
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (E.M.T.); (E.C.); (S.P.); (I.G.); (B.C.)
| |
Collapse
|
5
|
Meyer FEU, Santos GL, Doan TP, DeGrave AN, Bues B, Lutz S. Pirfenidone affects human cardiac fibroblast proliferation and cell cycle activity in 2D cultures and engineered connective tissues. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1687-1699. [PMID: 36800014 PMCID: PMC10338590 DOI: 10.1007/s00210-023-02421-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023]
Abstract
The anti-fibrotic drug pirfenidone (PFD) is currently in clinical testing for the treatment of heart failure with preserved ejection fraction; however, its effects on human cardiac cells have not been fully investigated. Therefore, we aimed to characterize the impact of PFD on human cardiac fibroblasts (CF) in 2D culture as well as in 3D-engineered connective tissues (ECT). We analyzed proliferation by automated cell counting and changes in signaling by immunoblotting. We generated ECT with different geometries to modify the cellular phenotype and investigated the effects of PFD on cell number and viability as well as on cell cycle activity. We further studied its effect on ECT compaction, contraction, stiffening, and strain resistance by ECT imaging, pole deflection analysis, and ultimate tensile testing. Our data demonstrate that PFD inhibits human CF proliferation in a concentration-dependent manner with an IC50 of 0.43 mg/ml and its anti-mitogenic effect was further corroborated by an inhibition of MEK1/2, ERK1/2, and riboprotein S6 (rpS6) phosphorylation. In ECT, a lower cell cycle activity was found in PFD-treated ECT and fewer cells resided in these ECT after 5 days of culture compared to the control. Moreover, ECT compaction as well as ECT contraction was impaired. Consequently, biomechanical analyses demonstrated that PFD reduced the stiffness of ECT. Taken together, our data demonstrate that the anti-fibrotic action of PFD on human CF is based on its anti-mitogenic effect in 2D cultures and ECT.
Collapse
Affiliation(s)
| | - Gabriela Leao Santos
- Institute of Pharmacology and Toxicology, University Medical Center, Goettingen, Germany
- Randall Centre for Cell and Molecular Biophysics, Kings College London, London, UK
- DZHK (German Centre for Cardiovascular Research) Partner Site, Goettingen, Germany
| | - Thao Phuong Doan
- Institute of Pharmacology and Toxicology, University Medical Center, Goettingen, Germany
| | - Alisa Nicole DeGrave
- Institute of Pharmacology and Toxicology, University Medical Center, Goettingen, Germany
- DZHK (German Centre for Cardiovascular Research) Partner Site, Goettingen, Germany
| | - Bastian Bues
- Institute of Pharmacology and Toxicology, University Medical Center, Goettingen, Germany
| | - Susanne Lutz
- Institute of Pharmacology and Toxicology, University Medical Center, Goettingen, Germany.
- DZHK (German Centre for Cardiovascular Research) Partner Site, Goettingen, Germany.
| |
Collapse
|
6
|
Chung EP, Nguyen JQ, Tellkamp-Schehr T, Goebel K, Ollek A, Krein C, Wells AR, Sebastian EA, Goebel A, Niese S, Leung KP. A Soft Skin Adhesive (SSA) Patch for Extended Release of Pirfenidone in Burn Wounds. Pharmaceutics 2023; 15:1842. [PMID: 37514029 PMCID: PMC10386754 DOI: 10.3390/pharmaceutics15071842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
As much as half or more of deep partial-thickness burn wounds develop hypertrophic scarring and contracture. Once formed, treatments are only minimally effective. Pirfenidone (Pf), indicated for treatment of idiopathic pulmonary fibrosis, is an anti-inflammatory and anti-fibrotic small molecule that potentially can be repurposed as a preventative against scarring in burn wounds. We present a drug-in-matrix patch with a soft skin adhesive (SSA) wound-contacting layer for multi-day drug delivery of Pf into burn wounds at the point of injury. Our patch construction consists of an SSA adhesive layer (Liveo™ MG7-9850, Dupont, Wilmington, DE, USA) for wound fixation, an acrylic co-polymer drug matrix (DURO-TAK 87-2852, Henkel, Düsseldorf, Germany) as the drug (Pf) reservoir, and an outermost protective polyurethane backing. By employing a drug-in-matrix patch design, Pf can be loaded as high as 2 mg/cm2. Compared to the acrylic co-polymer adhesive patch preparations and commercial films, adding an SSA layer markedly reduces skin stripping observed under scanning electron microscopy (SEM). Moreover, the addition of varying SSA thicknesses did not interfere with the in vitro release kinetics or drug permeation in ex vivo porcine skin. The Pf patch can be easily applied onto and removed from deep partial-thickness burn wounds on Duroc pigs. Continuous multi-day dosing of Pf by the patches (>200 μg/cm2/day) reduced proinflammatory biomarkers in porcine burn wounds. Pf patches produced by the manual laboratory-scale process showed excellent stability, maintaining intact physical patch properties and in vitro biological activity for up to one year under long-term (25 °C at 60% RH) and 6 months under accelerated (40 °C at 75% RH) test conditions. To manufacture our wound safe-and-extended-release patch, we present scale-up processes using a machine-driven automated roll-to-roll pilot scale coater.
Collapse
Affiliation(s)
- Eugene P Chung
- Combat Wound Care Group, US Army Institute of Surgical Research, JBSA, Fort Sam Houston, TX 78234, USA
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Jesse Q Nguyen
- Combat Wound Care Group, US Army Institute of Surgical Research, JBSA, Fort Sam Houston, TX 78234, USA
| | | | - Katja Goebel
- Labtec GmbH, Raiffeisenstrasse 4, 40764 Langenfeld, Germany
| | - Anita Ollek
- Labtec GmbH, Raiffeisenstrasse 4, 40764 Langenfeld, Germany
| | - Cliff Krein
- Labtec GmbH, Raiffeisenstrasse 4, 40764 Langenfeld, Germany
| | - Adrienne R Wells
- Combat Wound Care Group, US Army Institute of Surgical Research, JBSA, Fort Sam Houston, TX 78234, USA
- MicRoN Core, Harvard Medical School, Boston, MA 02215, USA
| | - Eliza A Sebastian
- Combat Wound Care Group, US Army Institute of Surgical Research, JBSA, Fort Sam Houston, TX 78234, USA
| | - Anja Goebel
- Labtec GmbH, Raiffeisenstrasse 4, 40764 Langenfeld, Germany
| | - Svenja Niese
- Labtec GmbH, Raiffeisenstrasse 4, 40764 Langenfeld, Germany
| | - Kai P Leung
- Combat Wound Care Group, US Army Institute of Surgical Research, JBSA, Fort Sam Houston, TX 78234, USA
| |
Collapse
|
7
|
Lescoat A, Roofeh D, Kuwana M, Lafyatis R, Allanore Y, Khanna D. Therapeutic Approaches to Systemic Sclerosis: Recent Approvals and Future Candidate Therapies. Clin Rev Allergy Immunol 2023; 64:239-261. [PMID: 34468946 PMCID: PMC9034469 DOI: 10.1007/s12016-021-08891-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2021] [Indexed: 02/06/2023]
Abstract
Systemic sclerosis is the rheumatic disease with the highest individual mortality. The severity of the disease is determined by the extent of fibrotic changes to cutaneous and internal organ tissues, the most life-threatening visceral manifestations being interstitial lung disease, SSc-associated-pulmonary arterial hypertension and myocardial involvement. The heterogeneity of the disease has initially hindered the design of successful clinical trials, but considerations on classification criteria have improved patient selection in trials, allowing the identification of more homogeneous groups of patients based on progressive visceral manifestations or the extent of skin involvement with a focus of patients with early disease. Two major subsets of systemic sclerosis are classically described: limited cutaneous systemic sclerosis characterized by distal skin fibrosis and the diffuse subset with distal and proximal skin thickening. Beyond this dichotomic subgrouping of systemic sclerosis, new phenotypic considerations based on antibody subtypes have provided a better understanding of the heterogeneity of the disease, anti-Scl70 antibodies being associated with progressive interstitial lung disease regardless of cutaneous involvement. Two targeted therapies, tocilizumab (a monoclonal antibody targeting interleukin-6 receptors (IL-6R)) and nintedanib (a tyrosine kinase inhibitor), have recently been approved by the American Food & Drug Administration to limit the decline of lung function in patients with SSc-associated interstitial lung disease, demonstrating that such better understanding of the disease pathogenesis with the identification of key targets can lead to therapeutic advances in the management of some visceral manifestations of the disease. This review will provide a brief overview of the pathogenesis of SSc and will present a selection of therapies recently approved or evaluated in this context. Therapies evaluated and approved in SSc-ILD will be emphasized and a review of recent phase II trials in diffuse cutaneous systemic sclerosis will be proposed. We will also discuss selected therapeutic pathways currently under investigation in systemic sclerosis that still lack clinical data in this context but that may show promising results in the future based on preclinical data.
Collapse
Affiliation(s)
- Alain Lescoat
- Department of Internal Medicine and Clinical Immunology, Rennes University Hospital, Rennes, France
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, France
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Michigan Scleroderma Program, University of Michigan, Ann Arbor, MI, USA
| | - David Roofeh
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Michigan Scleroderma Program, University of Michigan, Ann Arbor, MI, USA
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yannick Allanore
- INSERM U1016 and CNRS UMR8104, Institut Cochin, Paris, France
- Université de Paris, Université Paris Descartes, Paris, France
- Service de Rhumatologie, Hôpital Cochin, AP-HP.CUP, Paris, France
| | - Dinesh Khanna
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
- Michigan Scleroderma Program, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Fertala J, Wang ML, Rivlin M, Beredjiklian PK, Abboud J, Arnold WV, Fertala A. Extracellular Targets to Reduce Excessive Scarring in Response to Tissue Injury. Biomolecules 2023; 13:biom13050758. [PMID: 37238628 DOI: 10.3390/biom13050758] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Excessive scar formation is a hallmark of localized and systemic fibrotic disorders. Despite extensive studies to define valid anti-fibrotic targets and develop effective therapeutics, progressive fibrosis remains a significant medical problem. Regardless of the injury type or location of wounded tissue, excessive production and accumulation of collagen-rich extracellular matrix is the common denominator of all fibrotic disorders. A long-standing dogma was that anti-fibrotic approaches should focus on overall intracellular processes that drive fibrotic scarring. Because of the poor outcomes of these approaches, scientific efforts now focus on regulating the extracellular components of fibrotic tissues. Crucial extracellular players include cellular receptors of matrix components, macromolecules that form the matrix architecture, auxiliary proteins that facilitate the formation of stiff scar tissue, matricellular proteins, and extracellular vesicles that modulate matrix homeostasis. This review summarizes studies targeting the extracellular aspects of fibrotic tissue synthesis, presents the rationale for these studies, and discusses the progress and limitations of current extracellular approaches to limit fibrotic healing.
Collapse
Affiliation(s)
- Jolanta Fertala
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mark L Wang
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Michael Rivlin
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Pedro K Beredjiklian
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Joseph Abboud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - William V Arnold
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Andrzej Fertala
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
9
|
Abstract
INTRODUCTION Benign scarring central airway stenosis can be managed by high-pressure balloon dilatation, laser, surgery and stent implantation. The stenosis may have a high recurrence rate that necessitates repeated treatment. Pirfenidone (PFD) has anti-fibrosis effects and has been used in a variety of fibrosis diseases. Animal experiments suggested that PFD can prevent tracheal stenosis. PATIENT CONCERNS Patients with scarring central airway stenosis usually have chest tightness, cough and dyspnea. DIAGNOSIS Computed tomography scanning showed stenosis of the trachea and/or bronchus. Bronchoscopy revealed occlusion or stenosis of the trachea or bronchus. INTERVENTIONS The use of PFD in combination with other interventional management was reported to treat 2 cases of tracheobronchial stenosis after injury in this study. In the combined use of PFD and interventional management, PFD could help to alleviate tracheobronchial stenosis, prolong the time interval of bronchoscopic interventional treatment, and reduce medical costs. OUTCOMES The stenosis in the trachea and/or bronchus is relieved and the patients do not have any relevant symptoms.
Collapse
Affiliation(s)
- Xiao Li
- Department of Respiratory and Critical Care Medicine, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, China
| | - Jinbing Pan
- Department of Respiratory and Critical Care Medicine, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, China
- * Correspondence: Jinbing Pan, Department of Respiratory and Critical Care Medicine, Henan Provincial People’s Hospital, Zhengzhou University, 7 Weiwu Road, Zhengzhou 450003, China (e-mail: )
| | - Haoyu Qian
- Department of Respiratory and Critical Care Medicine, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, China
| | - Yun Ma
- Department of Respiratory and Critical Care Medicine, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, China
| | - Bulang Gao
- Department of Respiratory and Critical Care Medicine, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
10
|
Cristodor PL, Nechifor A, Fotea S, Nadasdy T, Bahloul Y, Nicolescu AC, Tatu AL. New Antifibroblastic Medication in Dermatology: Could Nintedanib Treat Scarring? Int J Gen Med 2022; 15:7169-7172. [PMID: 36118185 PMCID: PMC9480593 DOI: 10.2147/ijgm.s377073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/23/2022] [Indexed: 11/23/2022] Open
Abstract
There are a wide variety of disfiguring dermatological conditions whose pathologic substrate is represented by the unwanted deposition of collagen from dermal fibroblasts. Pirfenidone has demonstrated efficiency in the treatment of disordered collagen production when applied topically. Due to a similar mechanism of action, we also hypothesize that a similar medication, nintedanib, might have similar applications. We also propose that a liposomal technology may assist in the penetration of nintedanib and enhance its clinical effects.
Collapse
Affiliation(s)
- Patricia Liana Cristodor
- Center for the Morphologic Study of the Skin MORPHODERM, University of Medicine and Pharmacy “Victor Babeș”, Timișoara, TM, Romania
- Dermatology Department, Spitalul Clinic Municipal de Urgenta Timisoara, Timişoara, TM, Romania
| | - Alexandru Nechifor
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Galati, GL, Romania
| | - Silvia Fotea
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Galati, GL, Romania
| | - Thomas Nadasdy
- Dermatology Department, Spitalul Clinic Municipal de Urgenta Timisoara, Timişoara, TM, Romania
- Multidisciplinary Integrative Center for Dermatologic Interface Research MIC-DIR, Galati, GL, Romania
- Correspondence: Thomas Nadasdy; Yousef Bahloul, Dermatology Department, Spitalul Clinic Municipal de Urgenta Timisoara, str. Ofcea nr.24, Timişoara, TM, 300558, Romania, Tel +40 751609000, Email ;
| | - Yousef Bahloul
- Dermatology Department, Spitalul Clinic Municipal de Urgenta Timisoara, Timişoara, TM, Romania
- PhD Studies Department, University of Medicine and Pharmacy, Victor Babeș” Timișoara, Timișoara, TM, Romania
| | - Alin Codrut Nicolescu
- Department of Dermatology, ‘Roma’ Medical Center for Diagnosis and Treatment, Bucharest, Romania
| | - Alin Laurentiu Tatu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, Galati, GL, Romania
- Multidisciplinary Integrative Center for Dermatologic Interface Research MIC-DIR, Galati, GL, Romania
- Dermatology Department, “Sf. Cuvioasa Parascheva” Clinical Hospital of Infectious Diseases, Galati, GL, Romania
- Research Center in the Field of Medical and Pharmaceutical Sciences ReFORM-UDJ, Galati, GL, Romania
| |
Collapse
|
11
|
Bell TJ, Nagel DJ, Woeller CF, Kottmann RM. Ogerin mediated inhibition of TGF-β(1) induced myofibroblast differentiation is potentiated by acidic pH. PLoS One 2022; 17:e0271608. [PMID: 35901086 PMCID: PMC9333254 DOI: 10.1371/journal.pone.0271608] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 07/01/2022] [Indexed: 01/28/2023] Open
Abstract
Transforming growth factor beta (TGF-β) induced myofibroblast differentiation is central to the pathological scarring observed in Idiopathic Pulmonary Fibrosis (IPF) and other fibrotic diseases. Our lab has recently identified expression of GPR68 (Ovarian Cancer Gene Receptor 1, OGR1), a pH sensing G-protein coupled receptor, as a negative regulator of TGF-β induced profibrotic effects in primary human lung fibroblasts (PHLFs). We therefore hypothesized that small molecule activators of GPR68 would inhibit myofibroblast differentiation. Ogerin is a positive allosteric modulator (PAM) of GPR68, inducing a leftward shift of the dose response curve to proton induced signaling. Using PHLFs derived from patients with both non-fibrotic and IPF diagnoses, we show that Ogerin inhibits, and partially reverses TGF-β induced myofibroblast differentiation in a dose dependent manner. This occurs at the transcriptional level without inhibition of canonical TGF-β induced SMAD signaling. Ogerin induces PKA dependent CREB phosphorylation, a marker of Gαs pathway activation. The ability of Ogerin to inhibit both basal and TGF-β induced collagen gene transcription, and induction of Gαs signaling is enhanced at an acidic pH (pH 6.8). Similar findings were also found using fibroblasts derived from dermal, intestinal, and orbital tissue. The biological role of GPR68 in different tissues, cell types, and disease states is an evolving and emerging field. This work adds to the understanding of Gαs coupled GPCRs in fibrotic lung disease, the ability to harness the pH sensing properties of GPR68, and conserved mechanisms of fibrosis across different organ systems.
Collapse
Affiliation(s)
- Tyler J. Bell
- Department of Environmental Medicine Toxicology Training Program, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| | - David J. Nagel
- Department of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| | - Collynn F. Woeller
- Department of Ophthalmology, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
| | - R. Mathew Kottmann
- Department of Pulmonary and Critical Care Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States of America
- * E-mail:
| |
Collapse
|
12
|
Razia S, Park H, Shin E, Shim KS, Cho E, Kang MC, Kim SY. Synergistic effect of Aloe vera flower and Aloe gel on cutaneous wound healing targeting MFAP4 and its associated signaling pathway: In-vitro study. JOURNAL OF ETHNOPHARMACOLOGY 2022; 290:115096. [PMID: 35182666 DOI: 10.1016/j.jep.2022.115096] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/30/2022] [Accepted: 02/07/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aloe vera (L.) Burm. f. (Liliaceae family) is a well-known traditional medicinal plant, that has been used to treat a variety of illnesses, for decades ranging from cancer to skin disorders including wounds. It has been included in the traditional and herbal healthcare systems of many cultures around the world, as well as the pharmacopeia of different countries. Several in vitro and in vivo studies have also confirmed its potential antioxidant, anti-inflammatory, and wound-healing activities, etc. in the consistency of its historical and traditional uses. However, most studies to date are based on the A. vera gel and latex including its wound-healing effects. Very few studies have been focused on its flower, and rarely with its effects on cutaneous wound healing and its molecular mechanism. AIM OF THE STUDY To the best of our knowledge, this is the first study to report on the synergistic effect of the A. vera flower (AVF) and Aloe gel (PAG) on cutaneous wound-healing, as well as revealing its molecular mechanism targeting microfibril-associated glycoprotein 4 (MFAP4) and its associated signaling pathway. METHODS To investigate the synergistic effect of A. vera flower and Aloe gel in cutaneous wound healing, cell viability, and cell migration, as well proliferation assay was performed. This was followed by quantitative real-time polymerase chain reaction and Western blot analyses in wounded conditions to check the effects of this mixture on protein and mRNA levels in normal human dermal fibroblast (NHDF) cells. Moreover, small interfering RNA (siRNA) -mediated knockdown of MFAP4 in NHDF cells was performed followed by migration assay and cell cycle analysis, to confirm its role in cutaneous wound healing. Additionally, HaCaT cells were included in this study to evaluate its migratory and anti-inflammatory effects. RESULTS Based on our obtained results, the PAG and AVF mixture synergistically induced the proliferation, migration, and especially ECM formation of NHDF cells by enhancing the expression of MFAP4. Other extracellular components associated with MFAP4 signaling pathway, such as fibrillin, collagen, elastin, TGF β, and α-SMA, also increased at both the protein and mRNA levels. Subsequently, this mixture initiated the phosphorylation of the extracellular signal-regulated kinase (ERK) and AKT signaling pathways, and the S-phase of the cell cycle was also slightly modified. Also, the mixture induced the migration of HaCaT cells along with the suppression of inflammatory cytokines. Moreover, the siRNA-mediated knockdown highlighted the crucial role of MFAP4 in cutaneous wound healing in NHDF cells. CONCLUSION This study showed that the mixture of PAG and AVF has significant wound healing effects targeting MFAP4 and its associated signaling pathway. Additionally, MFAP4 was recognized as a new potential biomarker of wound healing, which can be confirmed by further in vivo studies.
Collapse
Affiliation(s)
- Sultana Razia
- Department of Life Science, University of Seoul, Seoul, 02504, South Korea; Department of Pharmacy, Jagannath University, Dhaka, 1100, Bangladesh
| | - Hyunsung Park
- Department of Life Science, University of Seoul, Seoul, 02504, South Korea
| | - Eunju Shin
- Univera Co., Ltd., Seoul, 04782, Republic of Korea
| | - Kyu-Suk Shim
- Univera Co., Ltd., Seoul, 04782, Republic of Korea
| | - Eunae Cho
- Univera Co., Ltd., Seoul, 04782, Republic of Korea
| | - Min Chol Kang
- College of Pharmacy, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon, 21936, South Korea
| | - Sun Yeou Kim
- College of Pharmacy, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon, 21936, South Korea.
| |
Collapse
|
13
|
Layer-by-Layer Pirfenidone/Cerium Oxide Nanocapsule Dressing Promotes Wound Repair and Prevents Scar Formation. Molecules 2022; 27:molecules27061830. [PMID: 35335197 PMCID: PMC8955702 DOI: 10.3390/molecules27061830] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 02/04/2023] Open
Abstract
An increase in the levels of reactive oxygen species (ROS) and high expression levels of transforming growth factor-β (TGF-β) in wound tissue are two major problems for wound repair and scar inhibition. Modulation of the wound microenvironment is considered to be able to overcome these issues. Two possible solutions include the use of cerium oxide nanoparticles (CeO2) as an enzyme-like ROS scavenger and pirfenidone (PFD) as an anti-fibrotic drug to inhibit the expression of TGF-β. However, CeO2 is easily adsorbed by biological macromolecules and loses its enzyme-like activity. Furthermore, the intracellular delivery of PFD is difficult. Herein, the layer-by-layer method was used to prepare nanocapsules (NCs) with a sophisticated structure featuring PFD at their core and CeO2 in their shell; these NCs were referred to as PFD/CeO2 NCs. PFD/CeO2 NCs were supposed to efficiently achieve intracellular delivery of PFD and successfully scavenged ROS from the microenvironment. Cellular experiments verified that PFD/CeO2 NCs had good biocompatibility, satisfactory cellular uptake, and favorable ROS-scavenging capacity. To be applied directly to the wound, PFD/CeO2 NCs were then adhered to plasma-etched polylactic acid (PLA) fiber membranes to prepare a new wound dressing. Animal experiments further demonstrated that the dressing accelerated the epithelialization of the wound, reduced the levels of ROS and TGF-β, improved the arrangement and proportion of collagen fibers, and finally, achieved satisfactory wound-repairing and anti-scarring effects. These results provide a new concept for promoting wound repair and preventing scar formation.
Collapse
|
14
|
Coentro JQ, May U, Prince S, Zwaagstra J, Ritvos O, Järvinen TAH, Zeugolis DI. Adapting the Scar-in-a-Jar to Skin Fibrosis and Screening Traditional and Contemporary Anti-Fibrotic Therapies. Front Bioeng Biotechnol 2021; 9:756399. [PMID: 34765594 PMCID: PMC8576412 DOI: 10.3389/fbioe.2021.756399] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/11/2021] [Indexed: 12/14/2022] Open
Abstract
Skin fibrosis still constitutes an unmet clinical need. Although pharmacological strategies are at the forefront of scientific and technological research and innovation, their clinical translation is hindered by the poor predictive capacity of the currently available in vitro fibrosis models. Indeed, customarily utilised in vitro scarring models are conducted in a low extracellular matrix milieu, which constitutes an oxymoron for the in-hand pathophysiology. Herein, we coupled macromolecular crowding (enhances and accelerates extracellular matrix deposition) with transforming growth factor β1 (TGFβ1; induces trans-differentiation of fibroblasts to myofibroblasts) in human dermal fibroblast cultures to develop a skin fibrosis in vitro model and to screen a range of anti-fibrotic families (corticosteroids, inhibitors of histone deacetylases, inhibitors of collagen crosslinking, inhibitors of TGFβ1 and pleiotropic inhibitors of fibrotic activation). Data obtained demonstrated that macromolecular crowding combined with TGFβ1 significantly enhanced collagen deposition and myofibroblast transformation. Among the anti-fibrotic compounds assessed, trichostatin A (inhibitors of histone deacetylases); serelaxin and pirfenidone (pleiotropic inhibitors of fibrotic activation); and soluble TGFβ receptor trap (inhibitor of TGFβ signalling) resulted in the highest decrease of collagen type I deposition (even higher than triamcinolone acetonide, the gold standard in clinical practice). This study further advocates the potential of macromolecular crowding in the development of in vitro pathophysiology models.
Collapse
Affiliation(s)
- João Q Coentro
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Ulrike May
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Stuart Prince
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - John Zwaagstra
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | | | - Tero A H Järvinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Tampere University Hospital, Tampere, Finland
| | - Dimitrios I Zeugolis
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway (NUI Galway), Galway, Ireland.,Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research and School of Mechanical and Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
15
|
Wu SB, Hou TY, Kau HC, Tsai CC. Effect of Pirfenidone on TGF-β1-Induced Myofibroblast Differentiation and Extracellular Matrix Homeostasis of Human Orbital Fibroblasts in Graves' Ophthalmopathy. Biomolecules 2021; 11:biom11101424. [PMID: 34680057 PMCID: PMC8533421 DOI: 10.3390/biom11101424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 01/01/2023] Open
Abstract
Pirfenidone is a pyridinone derivative that has been shown to inhibit fibrosis in animal models and in patients with idiopathic pulmonary fibrosis. Its effect on orbital fibroblasts remains poorly understood. We investigated the in vitro effect of pirfenidone in transforming growth factor-β1 (TGF-β1)-induced myofibroblast transdifferentiation and extracellular matrix (ECM) homeostasis in primary cultured orbital fibroblasts from patients with Graves' ophthalmopathy (GO). The expression of fibrotic proteins, including α-smooth muscle actin (α-SMA), connective tissue growth factor (CTGF), fibronectin, and collagen type I, was determined by Western blots. The activities of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) responsible for the ECM homeostasis were examined. After pretreating the GO orbital fibroblasts with pirfenidone (250, 500, and 750 μg/mL, respectively) for one hour followed by TGF-β1 for another 24 h, the expression of α-SMA, CTGF, fibronectin, and collagen type I decreased in a dose-dependent manner. Pretreating the GO orbital fibroblasts with pirfenidone not only abolished TGF-β1-induced TIMP-1 expression but recovered the MMP-2/-9 activities. Notably, pirfenidone inhibited TGF-β1-induced phosphorylation of p38 and c-Jun N-terminal kinase (JNK), the critical mediators in the TGF-β1 pathways. These findings suggest that pirfenidone modulates TGF-β1-mediated myofibroblast differentiation and ECM homeostasis by attenuating downstream signaling of TGF-β1.
Collapse
Affiliation(s)
- Shi-Bei Wu
- Biomedical Commercialization Center, Taipei Medical University, Taipei 11031, Taiwan;
| | - Tzu-Yu Hou
- Department of Ophthalmology, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan;
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
- School of Medicine, National Yang Ming University, Taipei 11221, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
| | - Hui-Chuan Kau
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
- School of Medicine, National Yang Ming University, Taipei 11221, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
- Department of Ophthalmology, Koo Foundation Sun Yat-Sen Cancer Center, Taipei 11259, Taiwan
| | - Chieh-Chih Tsai
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
- School of Medicine, National Yang Ming University, Taipei 11221, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
- Correspondence: ; Tel.: +886-2-28757325; Fax: +886-2-28213984
| |
Collapse
|
16
|
Lescoat A, Varga J, Matucci-Cerinic M, Khanna D. New promising drugs for the treatment of systemic sclerosis: pathogenic considerations, enhanced classifications, and personalized medicine. Expert Opin Investig Drugs 2021; 30:635-652. [PMID: 33909517 PMCID: PMC8292968 DOI: 10.1080/13543784.2021.1923693] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023]
Abstract
Introduction: Systemic sclerosis (SSc), also known as scleroderma, is a complex orphan disease characterized by early inflammatory features, vascular hyper-reactivity, and fibrosis of the skin and internal organs. Although substantial progress has been made in the understanding of the pathogenesis of SSc, there is still no disease-modifying drug that could significantly impact the natural history of the disease.Areas covered: This review discusses the rationale, preclinical evidence, first clinical eevidence,and pending issues concerning new promising therapeutic options that are under investigation in SSc. The search strategy was based on PubMed database and clinical trial.gov, highlighting recent key pathogenic aspects and phase I or II trials of investigational drugs in SSc.Expert opinion: The identification of new molecular entities that potentially impact inflammation and fibrosis may constitute promising options for a disease modifying-agent in SSc. The early combinations of antifibrotic drugs (such as pirfenidone) with immunomodulatory agents (such as mycophenolate mofetil) may also participate to achieve such a goal. A more refined stratification of patients, based on clinical features, molecular signatures, and identification of subpopulations with distinct clinical trajectories, may also improve management strategies in the future.
Collapse
Affiliation(s)
- Alain Lescoat
- Department of Internal Medicine and Clinical Immunology, Rennes University Hospital, Rennes, France
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, France
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Michigan Scleroderma Program, University of Michigan, Ann Arbor, Michigan, USA
| | - John Varga
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Michigan Scleroderma Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence, Italy
| | - Dinesh Khanna
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Michigan Scleroderma Program, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
17
|
Wei Q, Kong N, Liu X, Tian R, Jiao M, Li Y, Guan H, Wang K, Yang P. Pirfenidone attenuates synovial fibrosis and postpones the progression of osteoarthritis by anti-fibrotic and anti-inflammatory properties in vivo and in vitro. J Transl Med 2021; 19:157. [PMID: 33874948 PMCID: PMC8054406 DOI: 10.1186/s12967-021-02823-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/09/2021] [Indexed: 12/11/2022] Open
Abstract
Background Osteoarthritis (OA) is a disease of the entire joint involving synovial fibrosis and inflammation. Pathological changes to the synovium can accelerate the progression of OA. Pirfenidone (PFD) is a potent anti-fibrotic drug with additional anti-inflammatory properties. However, the influence of PFD on OA is unknown. Methods Proliferation of human fibroblast-like synoviocytes (FLSs) after treatment with TGF-β1 or PFD was evaluated using a Cell Counting Kit-8 assay and their migration using a Transwell assay. The expression of fibrosis-related genes (COL1A1, TIMP-1, and ACTA-2) and those related to inflammation (IL-6 and TNF-α) was quantified by real-time quantitative PCR. The protein expression levels of COL1A1, α-SMA (coded by ACTA-2), IL-6 and TNF-α were measured by enzyme-linked immunosorbent assay. A rabbit model of OA was established and then PFD was administered by gavage. The expression of genes related to fibrosis (COL1A1, TIMP-1, and ADAM-12) and inflammation (IL-6 and TNF-α) was measured using RNA extracted from the synovium. Synovial tissue was examined histologically after staining with H&E, Masson’s trichrome, and immunofluorescence. Synovitis scores, the volume fraction of collagen, and mean fluorescence intensity were calculated. Degeneration of articular cartilage was analyzed using a Safranin O-fast green stain and OARSI grading. Results The proliferation of FLSs was greatest when induced with 2.5 ng/ml TGF-β1 although it did not promote their migration. Therefore, 2.5 ng/ml TGF-β1 was used to stimulate the FLSs and evaluate the effects of PFD, which inhibited the migration of FLSs at concentrations as low as 1.0 mg/ml. PFD decreased the expression of COL1A1 while TGF-β1 increased both mRNA and protein expression levels of IL-6 but had no effect on α-SMA or TNF-α expression. PFD decreased mRNA expression levels of COL1A1, IL-6, and TNF-α in vivo. H&E staining and synovitis scores indicated that PFD reduced synovial inflammation, while Masson’s trichrome and immunofluorescence staining suggested that PFD decreased synovial fibrosis. Safranin O-Fast Green staining and the OARSI scores demonstrated that PFD delayed the progression of OA. Conclusions PFD attenuated synovial fibrosis and inflammation, and postponed the progression of osteoarthritis in a modified Hulth model of OA in rabbits, which was related to its anti-fibrotic and anti-inflammatory properties.
Collapse
Affiliation(s)
- Qilu Wei
- Bone and Joint Surgery Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Ning Kong
- Bone and Joint Surgery Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Xiaohui Liu
- Bone and Joint Surgery Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Run Tian
- Bone and Joint Surgery Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Ming Jiao
- Bone and Joint Surgery Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Yiyang Li
- Bone and Joint Surgery Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Huanshuai Guan
- Bone and Joint Surgery Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Kunzheng Wang
- Bone and Joint Surgery Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| | - Pei Yang
- Bone and Joint Surgery Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
18
|
De Pieri A, Korman BD, Jüngel A, Wuertz-Kozak K. Engineering Advanced In Vitro Models of Systemic Sclerosis for Drug Discovery and Development. Adv Biol (Weinh) 2021; 5:e2000168. [PMID: 33852183 PMCID: PMC8717409 DOI: 10.1002/adbi.202000168] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 12/19/2022]
Abstract
Systemic sclerosis (SSc) is a complex multisystem disease with the highest case-specific mortality among all autoimmune rheumatic diseases, yet without any available curative therapy. Therefore, the development of novel therapeutic antifibrotic strategies that effectively decrease skin and organ fibrosis is needed. Existing animal models are cost-intensive, laborious and do not recapitulate the full spectrum of the disease and thus commonly fail to predict human efficacy. Advanced in vitro models, which closely mimic critical aspects of the pathology, have emerged as valuable platforms to investigate novel pharmaceutical therapies for the treatment of SSc. This review focuses on recent advancements in the development of SSc in vitro models, sheds light onto biological (e.g., growth factors, cytokines, coculture systems), biochemical (e.g., hypoxia, reactive oxygen species) and biophysical (e.g., stiffness, topography, dimensionality) cues that have been utilized for the in vitro recapitulation of the SSc microenvironment, and highlights future perspectives for effective drug discovery and validation.
Collapse
Affiliation(s)
- Andrea De Pieri
- Dr. A. De Pieri, Prof. K. Wuertz-Kozak, Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY, 14623, USA
| | - Benjamin D Korman
- Prof. B. D. Korman, Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Astrid Jüngel
- Prof. A. Jüngel, Center of Experimental Rheumatology, University Clinic of Rheumatology, Balgrist University Hospital, University Hospital Zurich, Zurich, 8008, Switzerland
- Prof. A. Jüngel, Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Zurich, 8008, Switzerland
| | - Karin Wuertz-Kozak
- Dr. A. De Pieri, Prof. K. Wuertz-Kozak, Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY, 14623, USA
- Prof. K. Wuertz-Kozak, Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (Austria), Munich, 81547, Germany
| |
Collapse
|
19
|
Evani SJ, Karna SLR, Seshu J, Leung KP. Pirfenidone regulates LPS mediated activation of neutrophils. Sci Rep 2020; 10:19936. [PMID: 33203891 PMCID: PMC7672086 DOI: 10.1038/s41598-020-76271-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 09/24/2020] [Indexed: 12/16/2022] Open
Abstract
Excessive inflammation or its absence may result in impaired wound healing. Neutrophils are among the first innate immune cells to arrive at the injury site. They participate in infection control and debris removal to initiate healing. If not timely resolved, neutrophils can cause excessive tissue inflammation and damage. Drugs with anti-inflammatory and anti-fibrotic effects are of promise for improving healing by balancing the primary defensive functions and excessive tissue damage actions. Of interest, pirfenidone (Pf), an FDA approved anti-fibrotic drug to treat idiopathic pulmonary fibrosis, has been shown to ameliorate inflammation in several animal models including mouse deep partial-thickness burn wounds. However, there is a lack of mechanistic insights into Pf drug action on inflammatory cells such as neutrophils. Here, we examined the treatment effects of Pf on LPS-stimulated neutrophils as a model of non-sterile inflammation. Firstly, Pf reduced chemotaxis and production of pro-inflammatory ROS, cytokines, and chemokines by LPS-activated neutrophils. Secondly, Pf increased anti-inflammatory IL-1RA and reduced neutrophil degranulation, phagocytosis, and NETosis. Thirdly, Pf affected downstream signaling kinases which might directly or indirectly influence neutrophil responses to LPS. In conclusion, the results suggest that Pf lessens the inflammatory phenotypes of LPS-activated neutrophils.
Collapse
Affiliation(s)
- Shankar J Evani
- Division of Combat Wound Repair, U.S. Army Institute of Surgical Research, 3698 Chambers Pass, Building 3610, JBSA Fort Sam Houston, San Antonio, TX, 78234-7767, USA
| | - S L Rajasekhar Karna
- Division of Combat Wound Repair, U.S. Army Institute of Surgical Research, 3698 Chambers Pass, Building 3610, JBSA Fort Sam Houston, San Antonio, TX, 78234-7767, USA
| | - Janakiram Seshu
- South Texas Center for Emerging Infectious Diseases (STCEID) and Department of Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
| | - Kai P Leung
- Division of Combat Wound Repair, U.S. Army Institute of Surgical Research, 3698 Chambers Pass, Building 3610, JBSA Fort Sam Houston, San Antonio, TX, 78234-7767, USA.
| |
Collapse
|
20
|
Wilgus TA. Inflammation as an orchestrator of cutaneous scar formation: a review of the literature. PLASTIC AND AESTHETIC RESEARCH 2020; 7:54. [PMID: 33123623 PMCID: PMC7592345 DOI: 10.20517/2347-9264.2020.150] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inflammation is a key phase in the cutaneous wound repair process. The activation of inflammatory cells is critical for preventing infection in contaminated wounds and results in the release of an array of mediators, some of which stimulate the activity of keratinocytes, endothelial cells, and fibroblasts to aid in the repair process. However, there is an abundance of data suggesting that the strength of the inflammatory response early in the healing process correlates directly with the amount of scar tissue that will eventually form. This review will summarize the literature related to inflammation and cutaneous scar formation, highlight recent discoveries, and discuss potential treatment modalities that target inflammation to minimize scarring.
Collapse
Affiliation(s)
- Traci A Wilgus
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
21
|
Chung EP, Wells AR, Kiamco MM, Leung KP. Dual Asymmetric Centrifugation Efficiently Produces a Poloxamer-Based Nanoemulsion Gel for Topical Delivery of Pirfenidone. AAPS PharmSciTech 2020; 21:265. [PMID: 33006045 PMCID: PMC7529632 DOI: 10.1208/s12249-020-01798-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
This study used dual asymmetric centrifugation (DAC) to produce a topical vehicle for Pirfenidone (Pf; 5-methyl-1-phenyl-2[1H]-pyridone)—a Food and Drug Administration-approved antifibrotic drug indicated for idiopathic fibrosis treatment. Pf was loaded (8 wt%) in a poloxamer nanoemulsion gel (PNG) formulation consisting of water (47.8 wt%), triacetin (27.6 wt%), poloxamer 407 (P407, 13.8 wt%), polysorbate 80 (1.8 wt%), and benzyl alcohol (0.9 wt%). To our knowledge, poloxamer gels are typically processed with either high-shear methods or temperature regulation and have not been emulsified using DAC. Using a single-step emulsification process, 2 min mixed at 2500 RPM resulted in the lowest Pf loading variability with a relative standard deviation (RSD) of 0.96% for a 1.5 g batch size. Batch sizes of 15 g and 100 g yield higher RSD of 4.18% and 3.05%, respectively, but still in compliance with USP guidelines. Ex vivo permeation in full thickness porcine skin after 24 h showed total Pf permeation of 404.90 ± 67.07 μg/cm2. Tested in vitro on human dermal fibroblasts stimulated with transforming growth factor-beta 1 (TGF-β1), Pf-PNG resulted in a > 2 fold decrease in α-SMA expression over vehicle control demonstrating that formulated Pf retained its biological activity. One-month stability testing at 25°C/60% relative humidity (RH) and 40°C/75% RH showed that % drug content, release kinetics, and biological activity were largely unchanged for both conditions; however, pH decreased from 6.7 to 5.5 (25°C/60% RH) and 4.5 (40°C/75% RH) after 1 month. Overall, these data demonstrate the utility of DAC to rapidly and reproducibly prepare lab-scale batches of emulsified gels for pharmaceutical formulation development.
Collapse
|
22
|
Hsieh JC, Joshi CJ, Wan R, Galiano RD. The Northwestern Abdominoplasty Scar Model: A Tool for High-Throughput Assessment of Scar Therapeutics. Adv Wound Care (New Rochelle) 2020; 9:396-404. [PMID: 32320363 DOI: 10.1089/wound.2018.0900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Significance: Scar management is an important concern in plastic surgery. Scar models that best mimic in vivo human scarring are essential for understanding scar development and progression, assessing the efficacy of therapeutics, and providing reliable and valid research outcomes. Recent Advances: In 2016, Lanier et al. proposed a new in vivo patient model, the Northwestern Abdominoplasty Scar Model, that overcomes the prior limitations of both animal and human models, with greater representativeness of the human scarring process, expedited recruitment, smaller sample requirements, and greater flexibility in the types and number of interventions that can be studied simultaneously. Critical Issues: Existing animal models suffer from limitations that impede generalization to human scars. Human scar studies are difficult to conduct and rarely used due to recruitment difficulties, ethical concerns regarding purposeful wounding, and inherent variability based on location, type of scar, and the heterogeneity of the host response between humans. Although overcoming many of these hurdles, the Northwestern Abdominoplasty Scar Model still has a few limitations. In addition, there remains a need for further study of and comparison between the Northwestern Abdominoplasty Scar Model and existing human and animal models, to inspire more widespread acceptance of a standardized human scar model. Future Directions: The Northwestern Abdominoplasty Scar Model is a critical stepping stone toward better human scar models. This model hopefully will inspire other in vivo patient models utilizing elective surgery to overcome recruitment and ethical concerns.
Collapse
Affiliation(s)
- Ji-Cheng Hsieh
- Department of Plastic and Reconstructive Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Chitang J. Joshi
- Department of Plastic and Reconstructive Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Rou Wan
- Department of Plastic and Reconstructive Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Robert D. Galiano
- Department of Plastic and Reconstructive Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
23
|
Pirfenidone Inhibits Hypoxic Pulmonary Hypertension through the NADPH/ROS/p38 Pathway in Adventitial Fibroblasts in the Pulmonary Artery. Mediators Inflamm 2020; 2020:2604967. [PMID: 32587469 PMCID: PMC7305537 DOI: 10.1155/2020/2604967] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 05/22/2020] [Indexed: 02/08/2023] Open
Abstract
Hypoxic pulmonary hypertension (HPH) is a devastating disease characterized by progressive vasoconstriction and vascular remodeling. Pirfenidone (PFD) inhibits the progression of HPH, though the molecular mechanisms remain unknown. This study is aimed at determining the role and mechanism of PFD in HPH in human pulmonary artery adventitial fibroblasts (HPAAFs), which were cultured under normal or hypoxic conditions. NOX4 and Rac1 were inhibited or overexpressed by shRNA or pcDNA3.1, respectively. Proliferation of HPAAFs was quantified by colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) assays to assess cellular metabolic activity, cell counts, and ethynyldeoxyuridine (EdU) assays to detect DNA synthesis. Migration of HPAAFs was assessed by a wound healing assay. The expression levels of smooth muscle alpha-actin (a-SMA) and procollagen I (COL1A1) were assessed by RT-PCR and western blot analysis. PFD suppressed hypoxia-induced proliferation and migration of HPAAFs. Compared with the hypoxic control group, PFD reduced the expression of a-SMA and procollagen I (COL1A1). PFD reduced hypoxia-induced phosphorylation of p38 through the NOX4/reactive oxygen species (ROS) signaling pathway. Moreover, Rac1 also decreased hypoxia-induced phosphorylation of p38, without any cross-interaction with NOX4. These findings demonstrate that PFD is a novel therapeutic agent to prevent cell proliferation, migration, and fibrosis, which might be useful in inhibiting vascular remodeling in patients with HPH.
Collapse
|
24
|
Johnson BZ, Stevenson AW, Prêle CM, Fear MW, Wood FM. The Role of IL-6 in Skin Fibrosis and Cutaneous Wound Healing. Biomedicines 2020; 8:biomedicines8050101. [PMID: 32365896 PMCID: PMC7277690 DOI: 10.3390/biomedicines8050101] [Citation(s) in RCA: 234] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
The timely resolution of wound healing is critical for restoring the skin as a protective barrier. The switch from a proinflammatory to a reparative microenvironment must be tightly regulated. Interleukin (IL)-6 is a key modulator of the inflammatory and reparative process: it is involved in the differentiation, activation, and proliferation of leukocytes, endothelial cells, keratinocytes, and fibroblasts. This review examines the role of IL-6 in the healing of cutaneous wounds, and how dysregulation of IL-6 signaling can lead to either fibrosis or a failure to heal. The role of an IL-6/TGF-β feedback loop is discussed in the context of fibrogenesis, while IL-6 expression and responses in advanced age, diabetes, and obesity is outlined regarding the development of chronic wounds. Current research on therapies that modulate IL-6 is explored. Here, we consider IL-6′s diverse impact on cutaneous wound healing.
Collapse
Affiliation(s)
- Blair Z. Johnson
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (A.W.S.); (C.M.P.); (M.W.F.); (F.M.W.)
- Correspondence:
| | - Andrew W. Stevenson
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (A.W.S.); (C.M.P.); (M.W.F.); (F.M.W.)
| | - Cecilia M. Prêle
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (A.W.S.); (C.M.P.); (M.W.F.); (F.M.W.)
- Institute for Respiratory Health, University of Western Australia, Crawley, WA 6009, Australia
| | - Mark W. Fear
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (A.W.S.); (C.M.P.); (M.W.F.); (F.M.W.)
| | - Fiona M. Wood
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia; (A.W.S.); (C.M.P.); (M.W.F.); (F.M.W.)
- WA Department of Health, 189 Royal St, East Perth, WA 6004, Australia
| |
Collapse
|
25
|
Efficacy and Safety of Pirfenidone in Patients with Second-Degree Burns: A Proof-of-Concept Randomized Controlled Trial. Adv Skin Wound Care 2020; 33:1-7. [PMID: 32195729 DOI: 10.1097/01.asw.0000655484.95155.f7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVE Several studies suggest that pirfenidone may have a potential off-label use for wound healing. However, the effectiveness of this medication in patients with burns remains uncertain. Accordingly, investigators sought to assess wound re-epithelialization in patients with second-degree burns after adding pirfenidone to usual care. DESIGN AND SETTING Single-center pilot, proof-of-concept, single-blind randomized controlled trial. PATIENTS AND INTERVENTION Eight patients with second-degree burns were treated with occlusive hydrocolloid dressings and were randomly allocated to receive either no additional treatment or pirfenidone. OUTCOME MEASURES The primary outcome of the study was to evaluate wound healing between groups based on the thickness of the re-epithelialized epidermis at day 7. Secondary outcomes were to qualitatively assess the development of fibrotic tissue in the dermis, anomalies in the basal membrane, and the development of collagen fibers by histologic analysis. Liver and renal functions were measured daily to assess the overall safety of oral pirfenidone. MAIN RESULTS Patients treated with pirfenidone showed a remarkable improvement in wound re-epithelialization at day 7 (148.98 ± 13.64 vs 119.27 ± 15.55 μm; P = .029; 95% confidence interval, 4.14-55.29). Histologic evaluations showed less wound fibrosis in the pirfenidone group. CONCLUSIONS A decrease in wound healing time by enhancing wound re-epithelialization was observed with pirfenidone. Larger clinical trials are needed to reach more reliable conclusions.
Collapse
|
26
|
Wang XF, Fang QQ, Jia B, Hu YY, Wang ZC, Yan KP, Yin SY, Liu Z, Tan WQ. Potential effect of non-thermal plasma for the inhibition of scar formation: a preliminary report. Sci Rep 2020; 10:1064. [PMID: 31974451 PMCID: PMC6978306 DOI: 10.1038/s41598-020-57703-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/18/2019] [Indexed: 12/19/2022] Open
Abstract
Non-thermal plasma (NTP) is a promising biomedical tool for application to wound healing. However, there is limited scientific evidence that confirms its efficacy to inhibit scar formation. This study aims to investigate the role of non-thermal plasma in scar formation. Two full-thickness dorsal cutaneous wounds of rats were treated with either a non-thermal helium plasma jet or helium. It was determined that the non-thermal plasma jet accelerated the wound healing process from 5 days after surgery (day 5: 41.27% ± 2.351 vs 54.7% ± 5.314, p < 0.05; day 7: 56.05% ± 1.881 vs 75.28% ± 3.914, p < 0.01; day 14: 89.85% ± 2.991 vs 98.07% ± 0.839, p < 0.05). The width of the scars for the NTP group was narrower than those of control group (4.607 ± 0.416 mm vs 3.260 ± 0.333 mm, p < 0.05). In addition, a lower level of TGF-β1, p-Smad2 and p-Smad3 were detected in the NTP treated wounds (p < 0.05, p < 0.01 and p < 0.01). As expected, α-SMA was also significantly decreased in the NTP treatment group (p < 0.01). Moreover, the expression of type I collagen and the proportion of type I to III collagen were lower in the NTP group (p < 0.05). The results of the study suggest that NTP may play a potential role in scar formation by inhibiting the TGF β1 signal pathway and reducing the levels of α-SMA and type I collagen, and may have clinical utility in the future.
Collapse
Affiliation(s)
- Xiao-Feng Wang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, P.R. China
- Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, P.R. China
| | - Qing-Qing Fang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, P.R. China
- Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, P.R. China
| | - Bing Jia
- Institute of Industrial Ecology and Environment, Collage of Chemical and Biological Engineering, Zhejiang University, Zhejiang Province, P.R. China
| | - Yan-Yan Hu
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, P.R. China
- Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, P.R. China
| | - Zheng-Cai Wang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, P.R. China
- Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, P.R. China
| | - Ke-Ping Yan
- Institute of Industrial Ecology and Environment, Collage of Chemical and Biological Engineering, Zhejiang University, Zhejiang Province, P.R. China
| | - Sheng-Yong Yin
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, P.R. China
| | - Zhen Liu
- Institute of Industrial Ecology and Environment, Collage of Chemical and Biological Engineering, Zhejiang University, Zhejiang Province, P.R. China.
| | - Wei-Qiang Tan
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, P.R. China.
- Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, P.R. China.
| |
Collapse
|
27
|
Pirfenidone attenuates the profibrotic contractile phenotype of differentiated human dermal myofibroblasts. Biochem Biophys Res Commun 2019; 521:646-651. [PMID: 31679692 DOI: 10.1016/j.bbrc.2019.10.177] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022]
Abstract
Dysregulated wound healing after burn injury frequently results in debilitating hypertrophic scarring and contractures. Myofibroblasts, the main effector cells for dermal fibrosis, develop from normal fibroblasts via transforming growth factor beta 1 (TGF-β1). During wound healing, myofibroblasts produce extracellular matrix (ECM) proteins, modulate ECM stability, and contract the ECM using alpha smooth muscle actin (α-SMA) in contractile stress fibers. The antifibrotic pirfenidone has previously been shown to inhibit the initial differentiation of fibroblasts into myofibroblasts in vitro and act as a prophylactic measure against hypertrophic scar development in a mouse burn model. To test whether pirfenidone affects differentiated myofibroblasts, we investigated the in vitro effects of pirfenidone treatment after three to five days of stimulation with TGF-β1. In assays for morphology, protein and gene expression, and contractility, pirfenidone treatment produced significant effects. Profibrotic gene expression returned to near-normal levels, further α-SMA protein expression was prevented, and cell contraction within a stressed collagen matrix was reduced. These in vitro results promote pirfenidone as a promising antifibrotic agent to treat existing scars and healing wounds by mitigating the effects of differentiated myofibroblasts.
Collapse
|
28
|
Jin J, Togo S, Kadoya K, Tulafu M, Namba Y, Iwai M, Watanabe J, Nagahama K, Okabe T, Hidayat M, Kodama Y, Kitamura H, Ogura T, Kitamura N, Ikeo K, Sasaki S, Tominaga S, Takahashi K. Pirfenidone attenuates lung fibrotic fibroblast responses to transforming growth factor-β1. Respir Res 2019; 20:119. [PMID: 31185973 PMCID: PMC6558902 DOI: 10.1186/s12931-019-1093-z] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/05/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pirfenidone, an antifibrotic agent used for the treatment of idiopathic pulmonary fibrosis (IPF), functions by inhibiting myofibroblast differentiation, which is involved in transforming growth factor (TGF)-β1-induced IPF pathogenesis. However, unlike normal lung fibroblasts, the relationship between pirfenidone responses of TGF-β1-induced human fibrotic lung fibroblasts and lung fibrosis has not been elucidated. METHODS The effects of pirfenidone were evaluated in lung fibroblasts isolated from fibrotic human lung tissues after TGF-β1 exposure. The ability of two new pharmacological targets of pirfenidone, collagen triple helix repeat containing protein 1(CTHRC1) and four-and-a-half LIM domain protein 2 (FHL2), to mediate contraction of collagen gels and migration toward fibronectin were assessed in vitro. RESULTS Compared to control lung fibroblasts, pirfenidone significantly restored TGF-β1-stimulated fibroblast-mediated collagen gel contraction, migration, and CTHRC1 release in lung fibrotic fibroblasts. Furthermore, pirfenidone attenuated TGF-β1- and CTHRC1-induced fibroblast activity, upregulation of bone morphogenic protein-4(BMP-4)/Gremlin1, and downregulation of α-smooth muscle actin, fibronectin, and FHL2, similar to that observed post-CTHRC1 inhibition. In contrast, FHL2 inhibition suppressed migration and fibronectin expression, but did not downregulate CTHRC1. CONCLUSIONS Overall, pirfenidone suppressed fibrotic fibroblast-mediated fibrotic processes via inverse regulation of CTHRC1-induced lung fibroblast activity. Thus, CTHRC1 can be used for predicting pirfenidone response and developing new therapeutic targets for lung fibrosis.
Collapse
Affiliation(s)
- Jin Jin
- Department of Respiratory and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Beijing, 100730, People's Republic of China.,Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, 2-1 -1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Shinsaku Togo
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan. .,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, 2-1 -1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan. .,Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Kotaro Kadoya
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, 2-1 -1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Miniwan Tulafu
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, 2-1 -1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yukiko Namba
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, 2-1 -1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Department of Respiratory Medicine Kanagawa Cardiovascular and Respiratory Center, 6-16-1 Tomiokahigashi, Kanazawa-ku, Yokohama, Kanagawa, 236-0051, Japan
| | - Moe Iwai
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, 2-1 -1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Junko Watanabe
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, 2-1 -1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Kumi Nagahama
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, 2-1 -1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Takahiro Okabe
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, 2-1 -1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Moulid Hidayat
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, 2-1 -1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yuzo Kodama
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, 2-1 -1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hideya Kitamura
- Department of Respiratory Medicine Kanagawa Cardiovascular and Respiratory Center, 6-16-1 Tomiokahigashi, Kanazawa-ku, Yokohama, Kanagawa, 236-0051, Japan
| | - Takashi Ogura
- Department of Respiratory Medicine Kanagawa Cardiovascular and Respiratory Center, 6-16-1 Tomiokahigashi, Kanazawa-ku, Yokohama, Kanagawa, 236-0051, Japan
| | - Norikazu Kitamura
- Center for Information Biology, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540, Japan
| | - Kazuho Ikeo
- Center for Information Biology, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540, Japan.,Department of Genetics, SOKENDAI, 1111 Yata, Mishima, Shizuoka, 411-8540, Japan
| | - Shinichi Sasaki
- Department of Respiratory Medicine, Juntendo University Urayasu Hospital, Chiba, 279-0001, Japan
| | - Shigeru Tominaga
- Department of Respiratory Medicine, Juntendo University Urayasu Hospital, Chiba, 279-0001, Japan
| | - Kazuhisa Takahashi
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Research Institute for Diseases of Old Ages, Juntendo University Graduate School of Medicine, 2-1 -1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
29
|
Zhou C, Zeldin Y, Baratz ME, Kathju S, Satish L. Investigating the effects of Pirfenidone on TGF-β1 stimulated non-SMAD signaling pathways in Dupuytren's disease -derived fibroblasts. BMC Musculoskelet Disord 2019; 20:135. [PMID: 30927912 PMCID: PMC6441192 DOI: 10.1186/s12891-019-2486-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 03/03/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Dupuytren's disease (DD) is a progressive, debilitating condition of the hand that can eventually cause contractures of the affected fingers. Transforming growth factor- β1 (TGF-β1) has been reported to play a key role in DD pathology. Increased expression of TGF-β1 has shown to be the main stimulator of myofibroblast activity and in DD contractures. Pirfenidone (PFD), a small active molecule possess the ability to inhibit TGF-β1-mediated action in various fibrotic disorders. Our recent published findings show that PFD reduced TGF-β1-mediated cellular functions implicated in DD through SMAD signaling pathways. In the present study, the effect of PFD on TGF-β1-mediated non-SMAD signaling pathways were investigated in both carpal tunnel (CT) - and DD-derived fibroblasts. METHODS Fibroblasts harvested from Dupuytren's disease (DD) and carpal tunnel (CT) tissues were cultured in the presence or absence of TGF-β1 (10 ng/ml) and/or PFD (800 μg/ml). Cell lysates were analyzed using Western blots. Equal amounts of proteins were loaded to determine the phosphorylation levels of phosphatidylinositol-3 kinase (PI3K/AKT), extracellular regulated kinases (ERK1/2), p38 mitogen-activated protein kinase and Rho family related myosin light chain (MLC). RESULTS We show that the TGF-β1-induced phosphorylation of AKT was significantly decreased by the addition of PFD (800 μg/mL) in both CT- and DD-derived fibroblasts. Interestingly, there was no significant difference in the phosphorylation levels of both ERK and p38 on TGF-β1- induced cells in both CT-and DD-derived fibroblasts. But, PFD significantly decreased the TGF- β1-induced phosphorylation levels of ERK1/2 in both CT- and DD- cells. In contrast, PFD significantly decreased the basal and TGF- β1-induced phosphorylation levels of p38 in DD-derived fibroblasts. TGF- β1-induced phosphorylation levels of MLC was decreased by PFD in DD-derived fibroblasts. CONCLUSIONS These in-vitro results indicate for the first time that PFD has the potential to inhibit TGF-β1-induced non-SMAD signaling pathways in both CT- and DD-derived fibroblasts but pronounced statistically significant inhibition on all molecules was observed only in DD-derived fibroblasts. Our previous studies show that PFD can inhibit TGF-β1- induced SMAD signaling pathway proteins, namely p- SMAD2/SMAD3. These broad and complementary actions suggest PFD as a promising candidate to inhibit the TGF-β1- mediated molecular mechanisms leading to DD fibrosis.
Collapse
Affiliation(s)
- Chaoming Zhou
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261 USA
| | - Yael Zeldin
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261 USA
| | - Mark E. Baratz
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15261 USA
| | - Sandeep Kathju
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15219 USA
- Lumix Biomedical and Surgical Consulting, Pittsburgh, PA USA
| | - Latha Satish
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261 USA
- Shriners Hospitals for Children-Cincinnati, Cincinnati, OH 45229 USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati, 3229 Burnet Avenue, Cincinnati, OH 45229 USA
| |
Collapse
|
30
|
Türkmen E, Pata YS. Prevention of tracheal stenosis with pirfenidone after tracheotomy: An experimental study. Laryngoscope 2018; 129:E178-E186. [DOI: 10.1002/lary.27704] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/16/2018] [Accepted: 10/24/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Emre Türkmen
- Department of OtorhinolaryngologyIstanbul Medipol University Istanbul Turkey
| | - Yavuz Selim Pata
- Department of OtorhinolaryngologyYeditepe University Istanbul Turkey
| |
Collapse
|
31
|
Korfei M, Stelmaszek D, MacKenzie B, Skwarna S, Chillappagari S, Bach AC, Ruppert C, Saito S, Mahavadi P, Klepetko W, Fink L, Seeger W, Lasky JA, Pullamsetti SS, Krämer OH, Guenther A. Comparison of the antifibrotic effects of the pan-histone deacetylase-inhibitor panobinostat versus the IPF-drug pirfenidone in fibroblasts from patients with idiopathic pulmonary fibrosis. PLoS One 2018; 13:e0207915. [PMID: 30481203 PMCID: PMC6258535 DOI: 10.1371/journal.pone.0207915] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 11/08/2018] [Indexed: 12/20/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease with a poor prognosis. Pirfenidone is the first antifibrotic agent to be approved for IPF-treatment as it is able to slow down disease progression. However, there is no curative treatment other than lung transplantation. Because epigenetic alterations are associated with IPF, histone deacetylase (HDAC)-inhibitors have recently been proven to attenuate fibrotic remodeling in vitro and in vivo. This study compared the effects of pirfenidone with the pan-HDAC-inhibitor panobinostat/LBH589, a FDA-approved drug for the treatment of multiple myeloma, head-to-head on survival, fibrotic activity and proliferation of primary IPF-fibroblasts in vitro. Methods Primary fibroblasts from six IPF-patients were incubated for 24h with vehicle (0.25% DMSO), panobinostat (LBH589, 85 nM) or pirfenidone (2.7 mM), followed by assessment of proliferation and expression analyses for profibrotic and anti-apoptosis genes, as well as for ER stress and apoptosis-markers. In addition, the expression status of all HDAC enzymes was examined. Results Treatment of IPF-fibroblasts with panobinostat or pirfenidone resulted in a downregulated expression of various extracellular matrix (ECM)-associated genes, as compared to vehicle-treated cells. In agreement, both drugs decreased protein level of phosphorylated (p)-STAT3, a transcription factor mediating profibrotic responses, in treated IPF-fibroblasts. Further, an increase in histone acetylation was observed in response to both treatments, but was much more pronounced and excessive in panobinostat-treated IPF-fibroblasts. Panobinostat, but not pirfenidone, led to a significant suppression of proliferation in IPF-fibroblasts, as indicated by WST1- and BrdU assay and markedly diminished levels of cyclin-D1 and p-histone H3. Furthermore, panobinostat-treatment enhanced α-tubulin-acetylation, decreased the expression of survival-related genes Bcl-XL and BIRC5/survivin, and was associated with induction of ER stress and apoptosis in IPF-fibroblasts. In contrast, pirfenidone-treatment maintained Bcl-XL expression, and was neither associated with ER stress-induction nor any apoptotic signaling. Pirfenidone also led to increased expression of HDAC6 and sirtuin-2, and enhanced α-tubulin-deacetylation. But in line with its ability to increase histone acetylation, pirfenidone reduced the expression of HDAC enzymes HDAC1, -2 and -9. Conclusions We conclude that, beside other antifibrotic mechanisms, pirfenidone reduces profibrotic signaling also through STAT3 inactivation and weak epigenetic alterations in IPF-fibroblasts, and permits survival of (altered) fibroblasts. The pan-HDAC-inhibitor panobinostat reduces profibrotic phenotypes while inducing cell cycle arrest and apoptosis in IPF-fibroblasts, thus indicating more efficiency than pirfenidone in inactivating IPF-fibroblasts. We therefore believe that HDAC-inhibitors such as panobinostat can present a novel therapeutic strategy for IPF.
Collapse
Affiliation(s)
- Martina Korfei
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- * E-mail:
| | - Daniel Stelmaszek
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - BreAnne MacKenzie
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Sylwia Skwarna
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Shashipavan Chillappagari
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Anna C. Bach
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Clemens Ruppert
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Giessen, Germany
| | - Shigeki Saito
- Department of Medicine, Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Poornima Mahavadi
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Walter Klepetko
- Department of Thoracic Surgery, Vienna General Hospital, Vienna, Austria
- European IPF Network and European IPF Registry, Giessen, Germany
| | - Ludger Fink
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Giessen, Germany
- Institute of Pathology and Cytology, Wetzlar, Germany
| | - Werner Seeger
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Giessen, Germany
- Max-Planck-Institute for Heart and Lung Research, Department of Lung Development and Remodeling, Bad Nauheim, Germany
| | - Joseph A. Lasky
- Department of Medicine, Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Soni S. Pullamsetti
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Max-Planck-Institute for Heart and Lung Research, Department of Lung Development and Remodeling, Bad Nauheim, Germany
| | - Oliver H. Krämer
- Department of Toxicology, University Medical Center, Mainz, Germany
| | - Andreas Guenther
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Giessen, Germany
- European IPF Network and European IPF Registry, Giessen, Germany
- Agaplesion Lung Clinic Waldhof Elgershausen, Greifenstein, Germany
| |
Collapse
|
32
|
Pirfenidone Ointment Modulates the Burn Wound Bed in C57BL/6 Mice by Suppressing Inflammatory Responses. Inflammation 2018; 42:45-53. [DOI: 10.1007/s10753-018-0871-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
33
|
Kim Y, Park N, Rim YA, Nam Y, Jung H, Lee K, Ju JH. Establishment of a complex skin structure via layered co-culture of keratinocytes and fibroblasts derived from induced pluripotent stem cells. Stem Cell Res Ther 2018; 9:217. [PMID: 30103800 PMCID: PMC6090613 DOI: 10.1186/s13287-018-0958-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 12/11/2022] Open
Abstract
Background Skin is an organ that plays an important role as a physical barrier and has many other complex functions. Skin mimetics may be useful for studying the pathophysiology of diseases in vitro and for repairing lesions in vivo. Cord blood mononuclear cells (CBMCs) have emerged as a potential cell source for regenerative medicine. Human induced pluripotent stem cells (iPSCs) derived from CBMCs have great potential for allogenic regenerative medicine. Further study is needed on skin differentiation using CBMC-iPSCs. Methods Human iPSCs were generated from CBMCs by Sendai virus. CBMC-iPSCs were differentiated to fibroblasts and keratinocytes using embryonic body formation. To generate CBMC-iPSC-derived 3D skin organoid, CBMC-iPSC-derived fibroblasts were added into the insert of a Transwell plate and CBMC-iPSC-derived keratinocytes were seeded onto the fibroblast layer. Transplantation of 3D skin organoid was performed by the tie-over dressing method. Results Epidermal and dermal layers were developed using keratinocytes and fibroblasts differentiated from cord blood-derived human iPSCs, respectively. A complex 3D skin organoid was generated by overlaying the epidermal layer onto the dermal layer. A humanized skin model was generated by transplanting this human skin organoid into SCID mice and effectively healed skin lesions. Conclusions This study reveals that a human skin organoid generated using CBMC iPSCs is a novel tool for in-vitro and in-vivo dermatologic research. Electronic supplementary material The online version of this article (10.1186/s13287-018-0958-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yena Kim
- CiSTEM laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea
| | - Narae Park
- CiSTEM laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea
| | - Yeri Alice Rim
- CiSTEM laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea
| | - Yoojun Nam
- CiSTEM laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea
| | - Hyerin Jung
- CiSTEM laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea
| | - Kijun Lee
- CiSTEM laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea
| | - Ji Hyeon Ju
- CiSTEM laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea. .,Division of Rheumatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul, 137-701, Republic of Korea.
| |
Collapse
|
34
|
Dorati R, Medina JL, DeLuca PP, Leung KP. Development of a Topical 48-H Release Formulation as an Anti-scarring Treatment for Deep Partial-Thickness Burns. AAPS PharmSciTech 2018; 19:2264-2275. [PMID: 29790019 DOI: 10.1208/s12249-018-1030-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/23/2018] [Indexed: 02/08/2023] Open
Abstract
The purpose of this study was to develop pirfenidone (PF) ointment formulations for a dose finding study in the prophylactic treatment of deep partial-thickness burns in a mouse model. A preformulation study was performed to evaluate the solubility of PF in buffers and different solvents and its stability. Three different formulations containing 1, 3.5, and 6.5% w/w PF were prepared and optimized for their composition for testing in mice. Optimized formulations showed promising in vitro release profiles, in which 20-45% of PF was released in the first 7 h and 70-90% released within 48 h. The rheological properties of the ointment remained stable throughout storage at 25 ± 2°C/60% RH. Animal studies showed treatments of burn wounds during the inflammatory stage of wound healing with PF ointments at different drug concentrations had no adverse effects on reepithelization. Moreover, 6.5% PF ointment (F3) reduced the expression of pro-inflammatory cytokines IL-12p70 and TNFα. This study suggests that hydrocarbon base ointment could be a promising dosage form for topical delivery of PF in treatment of deep partial-thickness burns.
Collapse
|