1
|
Kadam AH, Schnitzer JE. Insights into Disease Progression of Translational Preclinical Rat Model of Interstitial Pulmonary Fibrosis through Endpoint Analysis. Cells 2024; 13:515. [PMID: 38534359 DOI: 10.3390/cells13060515] [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: 12/19/2023] [Revised: 02/06/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating interstitial lung disease characterized by the relentless deposition of extracellular matrix (ECM), causing lung distortions and dysfunction. Animal models of human IPF can provide great insight into the mechanistic pathways underlying disease progression and a means for evaluating novel therapeutic approaches. In this study, we describe the effect of bleomycin concentration on disease progression in the classical rat bleomycin model. In a dose-response study (1.5, 2, 2.5 U/kg i.t), we characterized lung fibrosis at day 14 after bleomycin challenge using endpoints including clinical signs, inflammatory cell infiltration, collagen content, and bronchoalveolar lavage fluid-soluble profibrotic mediators. Furthermore, we investigated fibrotic disease progression after 2 U/kg i.t. bleomycin administration at days 3, 7, and 14 by quantifying the expression of clinically relevant signaling molecules and pathways, epithelial mesenchymal transition (EMT) biomarkers, ECM components, and histopathology of the lung. A single bleomycin challenge resulted in a progressive fibrotic response in rat lung tissue over 14 days based on lung collagen content, histopathological changes, and modified Ashcroft score. The early fibrogenesis phase (days 3 to 7) is associated with an increase in profibrotic mediators including TGFβ1, IL6, TNFα, IL1β, CINC1, WISP1, VEGF, and TIMP1. In the mid and late fibrotic stages, the TGFβ/Smad and PDGF/AKT signaling pathways are involved, and clinically relevant proteins targeting galectin-3, LPA1, transglutaminase-2, and lysyl oxidase 2 are upregulated on days 7 and 14. Between days 7 and 14, the expressions of vimentin and α-SMA proteins increase, which is a sign of EMT activation. We confirmed ECM formation by increased expressions of procollagen-1Aα, procollagen-3Aα, fibronectin, and CTGF in the lung on days 7 and 14. Our data provide insights on a complex network of several soluble mediators, clinically relevant signaling pathways, and target proteins that contribute to drive the progressive fibrotic phenotype from the early to late phase (active) in the rat bleomycin model. The framework of endpoints of our study highlights the translational value for pharmacological interventions and mechanistic studies using this model.
Collapse
Affiliation(s)
- Anil H Kadam
- Proteogenomics Research Institute for Systems Medicine (PRISM), 505 Coast Blvd. South, La Jolla, CA 92037, USA
| | - Jan E Schnitzer
- Proteogenomics Research Institute for Systems Medicine (PRISM), 505 Coast Blvd. South, La Jolla, CA 92037, USA
| |
Collapse
|
2
|
Pang N, Laiva AL, Sulaiman NZ, Das P, O’Brien FJ, Keogh MB. Dual Glyoxalase-1 and β-Klotho Gene-Activated Scaffold Reduces Methylglyoxal and Reprograms Diabetic Adipose-Derived Stem Cells: Prospects in Improved Wound Healing. Pharmaceutics 2024; 16:265. [PMID: 38399319 PMCID: PMC10892312 DOI: 10.3390/pharmaceutics16020265] [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: 12/23/2023] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Tissue engineering approaches aim to provide biocompatible scaffold supports that allow healing to progress often in healthy tissue. In diabetic foot ulcers (DFUs), hyperglycemia impedes ulcer regeneration, due to complications involving accumulations of cellular methylglyoxal (MG), a key component of oxidated stress and premature cellular aging which further limits repair. In this study, we aim to reduce MG using a collagen-chondroitin sulfate gene-activated scaffold (GAS) containing the glyoxalase-1 gene (GLO-1) to scavenge MG and anti-fibrotic β-klotho to restore stem cell activity in diabetic adipose-derived stem cells (dADSCs). dADSCs were cultured on dual GAS constructs for 21 days in high-glucose media in vitro. Our results show that dADSCs cultured on dual GAS significantly reduced MG accumulation (-84%; p < 0.05) compared to the gene-free controls. Similar reductions in profibrotic proteins α-smooth muscle actin (-65%) and fibronectin (-76%; p < 0.05) were identified in dual GAS groups. Similar findings were observed in the expression of pro-scarring structural proteins collagen I (-62%), collagen IV (-70%) and collagen VII (-86%). A non-significant decrease in the expression of basement membrane protein E-cadherin (-59%) was noted; however, the dual GAS showed a significant increase in the expression of laminin (+300%). We conclude that dual GAS-containing Glo-1 and β-klotho had a synergistic MG detoxification and anti-fibrotic role in dADSC's. This may be beneficial to provide better wound healing in DFUs by controlling the diabetic environment and rejuvenating the diabetic stem cells towards improved wound healing.
Collapse
Affiliation(s)
- Nadia Pang
- Tissue Engineering Research Group—Bahrain, Royal College of Surgeons in Ireland, Adliya P.O. Box 15503, Bahrain; (N.P.); (N.Z.S.); (P.D.)
| | - Ashang L. Laiva
- Tissue Engineering Research Group—Bahrain, Royal College of Surgeons in Ireland, Adliya P.O. Box 15503, Bahrain; (N.P.); (N.Z.S.); (P.D.)
| | - Noof Z. Sulaiman
- Tissue Engineering Research Group—Bahrain, Royal College of Surgeons in Ireland, Adliya P.O. Box 15503, Bahrain; (N.P.); (N.Z.S.); (P.D.)
| | - Priya Das
- Tissue Engineering Research Group—Bahrain, Royal College of Surgeons in Ireland, Adliya P.O. Box 15503, Bahrain; (N.P.); (N.Z.S.); (P.D.)
| | - Fergal J. O’Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green, D02 YN77 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Michael B. Keogh
- Tissue Engineering Research Group—Bahrain, Royal College of Surgeons in Ireland, Adliya P.O. Box 15503, Bahrain; (N.P.); (N.Z.S.); (P.D.)
| |
Collapse
|
3
|
Reed EB, Orbeta S, Miao BA, Sitikov A, Chen B, Levitan I, Solway J, Mutlu GM, Fang Y, Mongin AA, Dulin NO. Anoctamin-1 is induced by TGF-β and contributes to lung myofibroblast differentiation. Am J Physiol Lung Cell Mol Physiol 2024; 326:L111-L123. [PMID: 38084409 PMCID: PMC11279757 DOI: 10.1152/ajplung.00155.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/07/2023] [Accepted: 11/29/2023] [Indexed: 12/26/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by progressive scarring of the lungs and resulting in deterioration in lung function. Transforming growth factor-β (TGF-β) is one of the most established drivers of fibrotic processes. TGF-β promotes the transformation of tissue fibroblasts to myofibroblasts, a key finding in the pathogenesis of pulmonary fibrosis. We report here that TGF-β robustly upregulates the expression of the calcium-activated chloride channel anoctamin-1 (ANO1) in human lung fibroblasts (HLFs) at mRNA and protein levels. ANO1 is readily detected in fibrotic areas of IPF lungs in the same area with smooth muscle α-actin (SMA)-positive myofibroblasts. TGF-β-induced myofibroblast differentiation (determined by the expression of SMA, collagen-1, and fibronectin) is significantly inhibited by a specific ANO1 inhibitor, T16Ainh-A01, or by siRNA-mediated ANO1 knockdown. T16Ainh-A01 and ANO1 siRNA attenuate profibrotic TGF-β signaling, including activation of RhoA pathway and AKT, without affecting initial Smad2 phosphorylation. Mechanistically, TGF-β treatment of HLFs results in a significant increase in intracellular chloride levels, which is prevented by T16Ainh-A01 or by ANO1 knockdown. The downstream mechanism involves the chloride-sensing "with-no-lysine (K)" kinase (WNK1). WNK1 siRNA significantly attenuates TGF-β-induced myofibroblast differentiation and signaling (RhoA pathway and AKT), whereas the WNK1 kinase inhibitor WNK463 is largely ineffective. Together, these data demonstrate that 1) ANO1 is a TGF-β-inducible chloride channel that contributes to increased intracellular chloride concentration in response to TGF-β; and 2) ANO1 mediates TGF-β-induced myofibroblast differentiation and fibrotic signaling in a manner dependent on WNK1 protein but independent of WNK1 kinase activity.NEW & NOTEWORTHY This study describes a novel mechanism of differentiation of human lung fibroblasts (HLFs) to myofibroblasts: the key process in the pathogenesis of pulmonary fibrosis. Transforming growth factor-β (TGF-β) drives the expression of calcium-activated chloride channel anoctmin-1 (ANO1) leading to an increase in intracellular levels of chloride. The latter recruits chloride-sensitive with-no-lysine (K) kinase (WNK1) to activate profibrotic RhoA and AKT signaling pathways, possibly through activation of mammalian target of rapamycin complex-2 (mTORC2), altogether promoting myofibroblast differentiation.
Collapse
Affiliation(s)
- Eleanor B Reed
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Shaina Orbeta
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, United States
| | - Bernadette A Miao
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Albert Sitikov
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Bohao Chen
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Irena Levitan
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Julian Solway
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Gökhan M Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Yun Fang
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Alexander A Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, United States
| | - Nickolai O Dulin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| |
Collapse
|
4
|
South AP, Laimer M, Gueye M, Sui JY, Eichenfield LF, Mellerio JE, Nyström A. Type VII Collagen Deficiency in the Oncogenesis of Cutaneous Squamous Cell Carcinoma in Dystrophic Epidermolysis Bullosa. J Invest Dermatol 2023; 143:2108-2119. [PMID: 37327859 DOI: 10.1016/j.jid.2023.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/11/2023] [Accepted: 05/17/2023] [Indexed: 06/18/2023]
Abstract
Dystrophic epidermolysis bullosa is a rare genetic skin disorder caused by COL7A1 sequence variations that result in type VII collagen deficits and cutaneous and extracutaneous manifestations. One serious complication of dystrophic epidermolysis bullosa is cutaneous squamous cell carcinoma, a leading driver of morbidity and mortality, especially among patients with recessive dystrophic epidermolysis bullosa. Type VII collagen deficits alter TGFβ signaling and evoke multiple other cutaneous squamous cell carcinoma progression-promoting activities within epidermal microenvironments. This review examines cutaneous squamous cell carcinoma pathophysiology in dystrophic epidermolysis bullosa with a focus on known oncogenesis pathways at play and explores the idea that therapeutic type VII collagen replacement may reduce cutaneous squamous cell carcinoma risk.
Collapse
Affiliation(s)
- Andrew P South
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| | - Martin Laimer
- Department of Dermatology and Allergology and EB House Austria, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | | | - Jennifer Y Sui
- Departments of Dermatology and Pediatrics, University of California San Diego School of Medicine, San Diego, California, USA; Division of Pediatric Dermatology, Rady Children's Hospital San Diego, San Diego, California, USA
| | - Lawrence F Eichenfield
- Departments of Dermatology and Pediatrics, University of California San Diego School of Medicine, San Diego, California, USA; Division of Pediatric Dermatology, Rady Children's Hospital San Diego, San Diego, California, USA
| | - Jemima E Mellerio
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center, University of Freiburg, Freiburg, Germany; Freiburg Institute for Advanced Studies, Freiburg, Germany
| |
Collapse
|
5
|
Siddiqui S, Bachert C, Bjermer L, Buchheit KM, Castro M, Qin Y, Rupani H, Sagara H, Howarth P, Taillé C. Eosinophils and tissue remodeling: Relevance to airway disease. J Allergy Clin Immunol 2023; 152:841-857. [PMID: 37343842 DOI: 10.1016/j.jaci.2023.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/15/2023] [Accepted: 06/02/2023] [Indexed: 06/23/2023]
Abstract
The ability of human tissue to reorganize and restore its existing structure underlies tissue homeostasis in the healthy airways, but in disease can persist without normal resolution, leading to an altered airway structure. Eosinophils play a cardinal role in airway remodeling both in health and disease, driving epithelial homeostasis and extracellular matrix turnover. Physiological consequences associated with eosinophil-driven remodeling include impaired lung function and reduced bronchodilator reversibility in asthma, and obstructed airflow in chronic rhinosinusitis with nasal polyps. Given the contribution of airway remodeling to the development and persistence of symptoms in airways disease, targeting remodeling is an important therapeutic consideration. Indeed, there is early evidence that eosinophil attenuation may reduce remodeling and disease progression in asthma. This review provides an overview of tissue remodeling in both health and airway disease with a particular focus on eosinophilic asthma and chronic rhinosinusitis with nasal polyps, as well as the role of eosinophils in these processes and the implications for therapeutic interventions. Areas for future research are also noted, to help improve our understanding of the homeostatic and pathological roles of eosinophils in tissue remodeling, which should aid the development of targeted and effective treatments for eosinophilic diseases of the airways.
Collapse
Affiliation(s)
- Salman Siddiqui
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| | - Claus Bachert
- Department of Otorhinolaryngology-Head and Neck Surgery, University Hospital of Münster, Münster, Germany; First Affiliated Hospital, Sun Yat-Sen University, International Airway Research Center, Guangzhou, China; Division of Ear, Nose, and Throat Diseases, Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden; Upper Airways Research Laboratory, Faculty of Medicine, Ghent University, Ghent, Belgium
| | - Leif Bjermer
- Department of Clinical Sciences, Respiratory Medicine, and Allergology, Lund University, Lund, Sweden
| | - Kathleen M Buchheit
- Jeff and Penny Vinik Center for Allergic Diseases Research, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Mario Castro
- Division of Pulmonary, Critical Care Medicine, University of Kansas School of Medicine, Kansas City, NC
| | - Yimin Qin
- Global Medical Affairs, Global Specialty and Primary Care, GlaxoSmithKline, Research Triangle Park, NC
| | - Hitasha Rupani
- Department of Respiratory Medicine, University Hospital Southampton National Health Service Foundation Trust, Southampton, United Kingdom
| | - Hironori Sagara
- Department of Medicine, Division of Respiratory Medicine and Allergology, Showa University, School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Peter Howarth
- Global Medical, Global Specialty and Primary Care, GlaxoSmithKline, Brentford, Middlesex, United Kingdom
| | - Camille Taillé
- Pneumology Department, Reference Center for Rare Pulmonary Diseases, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unit 1152, University of Paris Cité, Paris, France
| |
Collapse
|
6
|
Lee HJ, Tomasini-Johansson BR, Gupta N, Kwon GS. Fibronectin-targeted FUD and PEGylated FUD peptides for fibrotic diseases. J Control Release 2023; 360:69-81. [PMID: 37315694 PMCID: PMC10527082 DOI: 10.1016/j.jconrel.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023]
Abstract
Tissue fibrosis is characterized by excessive deposition of extracellular matrix (ECM) molecules. Fibronectin (FN) is a glycoprotein found in the blood and tissues, a key player in the assembly of ECM through interaction with cellular and extracellular components. Functional Upstream Domain (FUD), a peptide derived from an adhesin protein of bacteria, has a high binding affinity for the N-terminal 70-kDa domain of FN that plays a crucial role in FN polymerization. In this regard, FUD peptide has been characterized as a potent inhibitor of FN matrix assembly, reducing excessive ECM accumulation. Furthermore, PEGylated FUD was developed to prevent rapid elimination of FUD and enhance its systemic exposure in vivo. Herein, we summarize the development of FUD peptide as a potential anti-fibrotic agent and its application in experimental fibrotic diseases. In addition, we discuss how modification of the FUD peptide via PEGylation impacts pharmacokinetic profiles of the FUD peptide and can potentially contribute to anti-fibrosis therapy.
Collapse
Affiliation(s)
- Hye Jin Lee
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin - Madison, 777 Highland Avenue, Madison, WI 53705, USA
| | - Bianca R Tomasini-Johansson
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, 1111 Highland Avenue, WIMRII, Madison, WI 53705, USA
| | - Nikesh Gupta
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin - Madison, 777 Highland Avenue, Madison, WI 53705, USA
| | - Glen S Kwon
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin - Madison, 777 Highland Avenue, Madison, WI 53705, USA; Carbone Cancer Center, University of Wisconsin - Madison, 600 Highland Avenue, Madison, WI 53705, USA.
| |
Collapse
|
7
|
Heras‐Parets A, Ginebra M, Manero JM, Guillem‐Marti J. Guiding Fibroblast Activation Using an RGD-Mutated Heparin Binding II Fragment of Fibronectin for Gingival Titanium Integration. Adv Healthc Mater 2023; 12:e2203307. [PMID: 37100430 PMCID: PMC11468578 DOI: 10.1002/adhm.202203307] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/15/2023] [Indexed: 04/28/2023]
Abstract
The formation of a biological seal around the neck of titanium (Ti) implants is critical for ensuring integration at the gingival site and for preventing bacterial colonization that may lead to periimplantitis. This process is guided by activated fibroblasts, named myofibroblasts, which secrete extracellular matrix (ECM) proteins and ECM-degrading enzymes resolving the wound. However, in some cases, Ti is not able to attract and activate fibroblasts to a sufficient extent, which may compromise the success of the implant. Fibronectin (FN) is an ECM component found in wounds that is able to guide soft tissue healing through the adhesion of cells and attraction of growth factors (GFs). However, clinical use of FN functionalized Ti implants is problematic because FN is difficult to obtain, and is sensitive to degradation. Herein, functionalizing Ti with a modified recombinant heparin binding II (HBII) domain of FN, mutated to include an Arg-Gly-Asp (RGD) sequence for promoting both fibroblast adhesion and GF attraction, is aimed at. The HBII-RGD domain is able to stimulate fibroblast adhesion, spreading, proliferation, migration, and activation to a greater extent than the native HBII, reaching values closer to those of full-length FN suggesting that it might induce the formation of a biological sealing.
Collapse
Affiliation(s)
- Aina Heras‐Parets
- BiomaterialsBiomechanics and Tissue Engineering groupDepartment of Materials Science and EngineeringUniversitat Politècnica de Catalunya – BarcelonaTech (UPC)Av. Eduard Maristany 16Barcelona08930Spain
- Barcelona Research Center in Multiscale Science and EngineeringUPCAv. Eduard Maristany 16Barcelona08930Spain
| | - Maria‐Pau Ginebra
- BiomaterialsBiomechanics and Tissue Engineering groupDepartment of Materials Science and EngineeringUniversitat Politècnica de Catalunya – BarcelonaTech (UPC)Av. Eduard Maristany 16Barcelona08930Spain
- Barcelona Research Center in Multiscale Science and EngineeringUPCAv. Eduard Maristany 16Barcelona08930Spain
- Institute for Bioengineering of Catalonia (IBEC)Barcelona Institute of Science and Technology (BIST)Barcelona08028Spain
| | - Jose Maria Manero
- BiomaterialsBiomechanics and Tissue Engineering groupDepartment of Materials Science and EngineeringUniversitat Politècnica de Catalunya – BarcelonaTech (UPC)Av. Eduard Maristany 16Barcelona08930Spain
- Barcelona Research Center in Multiscale Science and EngineeringUPCAv. Eduard Maristany 16Barcelona08930Spain
| | - Jordi Guillem‐Marti
- BiomaterialsBiomechanics and Tissue Engineering groupDepartment of Materials Science and EngineeringUniversitat Politècnica de Catalunya – BarcelonaTech (UPC)Av. Eduard Maristany 16Barcelona08930Spain
- Barcelona Research Center in Multiscale Science and EngineeringUPCAv. Eduard Maristany 16Barcelona08930Spain
| |
Collapse
|
8
|
Baudo G, Wu S, Massaro M, Liu H, Lee H, Zhang A, Hamilton DJ, Blanco E. Polymer-Functionalized Mitochondrial Transplantation to Fibroblasts Counteracts a Pro-Fibrotic Phenotype. Int J Mol Sci 2023; 24:10913. [PMID: 37446100 PMCID: PMC10342003 DOI: 10.3390/ijms241310913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Fibroblast-to-myofibroblast transition (FMT) leads to excessive extracellular matrix (ECM) deposition-a well-known hallmark of fibrotic disease. Transforming growth factor-β (TGF-β) is the primary cytokine driving FMT, and this phenotypic conversion is associated with mitochondrial dysfunction, notably a metabolic reprogramming towards enhanced glycolysis. The objective of this study was to examine whether the establishment of favorable metabolic phenotypes in TGF-β-stimulated fibroblasts could attenuate FMT. The hypothesis was that mitochondrial replenishment of TGF-β-stimulated fibroblasts would counteract a shift towards glycolytic metabolism, consequently offsetting pro-fibrotic processes. Isolated mitochondria, functionalized with a dextran and triphenylphosphonium (TPP) (Dex-TPP) polymer conjugate, were administered to fibroblasts (MRC-5 cells) stimulated with TGF-β, and effects on bioenergetics and fibrotic programming were subsequently examined. Results demonstrate that TGF-β stimulation of fibroblasts led to FMT, which was associated with enhanced glycolysis. Dex-TPP-coated mitochondria (Dex-TPP/Mt) delivery to TGF-β-stimulated fibroblasts abrogated a metabolic shift towards glycolysis and led to a reduction in reactive oxygen species (ROS) generation. Importantly, TGF-β-stimulated fibroblasts treated with Dex-TPP/Mt had lessened expression of FMT markers and ECM proteins, as well as reduced migration and proliferation. Findings highlight the potential of mitochondrial transfer, as well as other strategies involving functional reinforcement of mitochondria, as viable therapeutic modalities in fibrosis.
Collapse
Affiliation(s)
- Gherardo Baudo
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Suhong Wu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Matteo Massaro
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haoran Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Hyunho Lee
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Aijun Zhang
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Dale J. Hamilton
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
- Department of Cardiology, Houston Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
9
|
Zhao T, Gong B, Luo S, Zhang R, Zhang L, Huang Y, Gao H, Gong T. A fibroblastic foci-targeting and hypoxia-cleavable delivery system of pirfenidone for the treatment of idiopathic pulmonary fibrosis. Acta Biomater 2023:S1742-7061(23)00349-5. [PMID: 37356783 DOI: 10.1016/j.actbio.2023.06.024] [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: 04/17/2023] [Revised: 05/31/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023]
Abstract
The progressive formation of fibroblastic foci characterizes idiopathic pulmonary fibrosis (IPF), and excessive oral doses of approved pirfenidone (PFD) always cause gastrointestinal side effects. The fibrotic response driven by activated fibroblasts could perpetuate epithelial damage and promote abnormal extracellular matrix (ECM) deposition. When modified nanoparticles reach their target, it is important to ensure a responsive release of PFD. Hypoxia is a determining factor in IPF, leading to alveolar dysfunction and deeper cellular fibrosis. Herein, a fibroblastic foci-targeting and hypoxia-cleavable drug delivery system (Fn-Azo-BSA@PEG) was established to reprogram the fibrosis in IPF. We have modified the FnBAP5 peptide to enable comprehensive fibroblastic foci targeting, which helps BSA nanoparticles recognize and accumulate at fibrotic sites. Meantime, the hypoxia-responsive azobenzene group allowed for efficient and rapid drug diffusion, while the PEGylated BSA reduced system toxicity and increased circulation in vivo. As expected, the strategy of the fibronectin-targeting-modification and hypoxia-responsive drug release synergistically inhibited activated fibroblasts and reduced the secretion of the fibrosis-related protein. Fn-Azo-BSA@PEG could accumulate in pulmonary tissue and prolong the survival time in bleomycin-induced pulmonary fibrosis mice. Together, the multivalent BSA nanoparticles offered an efficient approach for improving lung architecture and function by regulating the fibroblastic foci and hypoxia. STATEMENT OF SIGNIFICANCE: We established fibroblastic foci-targeting and hypoxia-cleavable bovine serum albumin (BSA) nanoparticles (Fn-Azo-BSA@PEG) to reprogramme the fibroblastic foci in idiopathic pulmonary fibrosis (IPF). Fn-Azo-BSA@PEG was designed to actively target fibroblasts and abnormal ECM with the FnBPA5 peptide, delivering more FDA-approved pirfenidone (PFD) to the cross-talk within the foci. Once the drug reached fibroblastic foci, the azobenzene group acted as a hypoxia-responsive linker to trigger effective and rapid drug release. Hypoxic responsiveness and FnBAP5-modification of Fn-Azo-BSA@PEG synergistically inhibited the secretion of proteins closely related to fibrogenesis. BSA's inherent transport and metabolic pathways in the pulmonary reduced the side effects of the main organs. The multivalent BSA nanoparticles efficiently inhibited IPF-fibrosis progress and preserved the lung architecture by regulating the fibroblastic foci and hypoxia.
Collapse
Affiliation(s)
- Ting Zhao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Bokai Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Shiqin Luo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Rongping Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Ling Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China.
| |
Collapse
|
10
|
Hume RD, Deshmukh T, Doan T, Shim WJ, Kanagalingam S, Tallapragada V, Rashid F, Marcuello M, Blessing D, Selvakumar D, Raguram K, Pathan F, Graham D, Ounzain S, Kizana E, Harvey RP, Palpant NJ, Chong JJ. PDGF-AB Reduces Myofibroblast Differentiation Without Increasing Proliferation After Myocardial Infarction. JACC Basic Transl Sci 2023. [DOI: 10.1016/j.jacbts.2022.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
|
11
|
Falcón-Cama V, Montero-González T, Acosta-Medina EF, Guillen-Nieto G, Berlanga-Acosta J, Fernández-Ortega C, Alfonso-Falcón A, Gilva-Rodríguez N, López-Nocedo L, Cremata-García D, Matos-Terrero M, Pentón-Rol G, Valdés I, Oramas-Díaz L, Suarez-Batista A, Noa-Romero E, Cruz-Sui O, Sánchez D, Borrego-Díaz AI, Valdés-Carreras JE, Vizcaino A, Suárez-Alba J, Valdés-Véliz R, Bergado G, González MA, Hernandez T, Alvarez-Arzola R, Ramírez-Suárez AC, Casillas-Casanova D, Lemos-Pérez G, Blanco-Águila OR, Díaz A, González Y, Bequet-Romero M, Marín-Prida J, Hernández-Perera JC, Del Rosario-Cruz L, Marin-Díaz AP, González-Bravo M, Borrajero I, Acosta-Rivero N. Evidence of SARS-CoV-2 infection in postmortem lung, kidney, and liver samples, revealing cellular targets involved in COVID-19 pathogenesis. Arch Virol 2023; 168:96. [PMID: 36842152 PMCID: PMC9968404 DOI: 10.1007/s00705-023-05711-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/29/2022] [Indexed: 02/27/2023]
Abstract
There is an urgent need to understand severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-host interactions involved in virus spread and pathogenesis, which might contribute to the identification of new therapeutic targets. In this study, we investigated the presence of SARS-CoV-2 in postmortem lung, kidney, and liver samples of patients who died with coronavirus disease (COVID-19) and its relationship with host factors involved in virus spread and pathogenesis, using microscopy-based methods. The cases analyzed showed advanced stages of diffuse acute alveolar damage and fibrosis. We identified the SARS-CoV-2 nucleocapsid (NC) in a variety of cells, colocalizing with mitochondrial proteins, lipid droplets (LDs), and key host proteins that have been implicated in inflammation, tissue repair, and the SARS-CoV-2 life cycle (vimentin, NLRP3, fibronectin, LC3B, DDX3X, and PPARγ), pointing to vimentin and LDs as platforms involved not only in the viral life cycle but also in inflammation and pathogenesis. SARS-CoV-2 isolated from a patient´s nasal swab was grown in cell culture and used to infect hamsters. Target cells identified in human tissue samples included lung epithelial and endothelial cells; lipogenic fibroblast-like cells (FLCs) showing features of lipofibroblasts such as activated PPARγ signaling and LDs; lung FLCs expressing fibronectin and vimentin and macrophages, both with evidence of NLRP3- and IL1β-induced responses; regulatory cells expressing immune-checkpoint proteins involved in lung repair responses and contributing to inflammatory responses in the lung; CD34+ liver endothelial cells and hepatocytes expressing vimentin; renal interstitial cells; and the juxtaglomerular apparatus. This suggests that SARS-CoV-2 may directly interfere with critical lung, renal, and liver functions involved in COVID-19-pathogenesis.
Collapse
Affiliation(s)
- Viviana Falcón-Cama
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba. .,Latin American School of Medicine, Calle Panamericana Km 3 1/2, Playa, 11600, Havana, Cuba.
| | | | - Emilio F Acosta-Medina
- Center for Advanced Studies of Cuba, Havana, Cuba. .,Latin American School of Medicine, Calle Panamericana Km 3 1/2, Playa, 11600, Havana, Cuba.
| | - Gerardo Guillen-Nieto
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba.,Latin American School of Medicine, Calle Panamericana Km 3 1/2, Playa, 11600, Havana, Cuba
| | - Jorge Berlanga-Acosta
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba.,Latin American School of Medicine, Calle Panamericana Km 3 1/2, Playa, 11600, Havana, Cuba
| | - Celia Fernández-Ortega
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba.,Latin American School of Medicine, Calle Panamericana Km 3 1/2, Playa, 11600, Havana, Cuba
| | | | - Nathalie Gilva-Rodríguez
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Lilianne López-Nocedo
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Daina Cremata-García
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Mariuska Matos-Terrero
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Giselle Pentón-Rol
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba.,Latin American School of Medicine, Calle Panamericana Km 3 1/2, Playa, 11600, Havana, Cuba
| | - Iris Valdés
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Leonardo Oramas-Díaz
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Anamarys Suarez-Batista
- Department of Virology, Civilian Defense Scientific Research Center (CICDC), Havana, Mayabeque, Cuba
| | - Enrique Noa-Romero
- Department of Virology, Civilian Defense Scientific Research Center (CICDC), Havana, Mayabeque, Cuba
| | - Otto Cruz-Sui
- Department of Virology, Civilian Defense Scientific Research Center (CICDC), Havana, Mayabeque, Cuba
| | | | | | | | | | - José Suárez-Alba
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Rodolfo Valdés-Véliz
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Gretchen Bergado
- Direction of Immunology and Immunotherapy, Center of Molecular Immunology, Havana, Cuba
| | - Miguel A González
- Direction of Immunology and Immunotherapy, Center of Molecular Immunology, Havana, Cuba
| | - Tays Hernandez
- Direction of Immunology and Immunotherapy, Center of Molecular Immunology, Havana, Cuba
| | - Rydell Alvarez-Arzola
- Direction of Immunology and Immunotherapy, Center of Molecular Immunology, Havana, Cuba
| | - Anna C Ramírez-Suárez
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Dionne Casillas-Casanova
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Gilda Lemos-Pérez
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | | | | | | | - Mónica Bequet-Romero
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Javier Marín-Prida
- Center for Research and Biological Evaluations, Institute of Pharmacy and Food, University of Havana, Havana, Cuba
| | | | | | - Alina P Marin-Díaz
- International Orthopedic Scientific Complex 'Frank Pais Garcia', Havana, Cuba
| | - Maritza González-Bravo
- Latin American School of Medicine, Calle Panamericana Km 3 1/2, Playa, 11600, Havana, Cuba
| | | | - Nelson Acosta-Rivero
- Center for Protein Studies, Department of Biochemistry, Faculty of Biology, University of Habana, Calle 25 entre J e I, #455, Plaza de la Revolucion, 10400, Havana, Cuba. .,Department of Infectious Diseases, Centre for Integrative Infectious Disease Research (CIID), Molecular Virology, University of Heidelberg, Medical Faculty Heidelberg, INF 344, GO.1, 69120, Heidelberg, Germany.
| |
Collapse
|
12
|
The Antibiotic Kitasamycin-A Potential Agent for Specific Fibrosis Preventing Therapy after Fistulating Glaucoma Surgery? Pharmaceutics 2023; 15:pharmaceutics15020329. [PMID: 36839651 PMCID: PMC9960401 DOI: 10.3390/pharmaceutics15020329] [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: 11/01/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023] Open
Abstract
One major complication after fistulating glaucoma surgeries are fibroblast-mediated scarring processes and their specific prevention is key in the development of novel pharmaceutical concepts. Within this study a possible antifibrotic potential of kitasamycin (KM) in a transforming growth factor (TGF)-β1-mediated fibroblast model was evaluated in vitro. Primary ocular fibroblasts were isolated, cultivated and a dose-response test including determination of the half maximal effective concentration (EC50) for KM was conducted. Transformation of fibroblasts into myofibroblasts was induced by TGF-β1and immunofluorescence (IF), and Western blot (WB) analyses were performed with fibroblasts and myofibroblasts. IF analyses were carried out using antibodies against α-smooth muscle actin (α-SMA) and fibronectin, and protein detection of intracellular and extracellular proteins was performed by WB. Using the dose-response test, the viability, cytotoxicity and EC50 of KM after 24 and 48 h were determined. Fibroblasts exposed to various KM concentrations showed no increase in α-SMA and extracellular matrix expression. In TGF-ß1-stimulated myofibroblasts, KM inhibited the expression of α-SMA and fibronectin in a concentration-dependent manner. These findings demonstrate that KM could impair the transformation of fibroblasts into myofibroblasts and the expression of proteins involved in fibrotic processes, representing a potential agent for specific fibrosis prevention in future therapeutic concepts.
Collapse
|
13
|
Atherton P, Konstantinou R, Neo SP, Wang E, Balloi E, Ptushkina M, Bennett H, Clark K, Gunaratne J, Critchley D, Barsukov I, Manser E, Ballestrem C. Tensin3 interaction with talin drives the formation of fibronectin-associated fibrillar adhesions. J Biophys Biochem Cytol 2022; 221:213452. [PMID: 36074065 PMCID: PMC9462884 DOI: 10.1083/jcb.202107022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 05/30/2022] [Accepted: 08/05/2022] [Indexed: 12/11/2022] Open
Abstract
The formation of healthy tissue involves continuous remodeling of the extracellular matrix (ECM). Whilst it is known that this requires integrin-associated cell-ECM adhesion sites (CMAs) and actomyosin-mediated forces, the underlying mechanisms remain unclear. Here, we examine how tensin3 contributes to the formation of fibrillar adhesions (FBs) and fibronectin fibrillogenesis. Using BioID mass spectrometry and a mitochondrial targeting assay, we establish that tensin3 associates with the mechanosensors such as talin and vinculin. We show that the talin R11 rod domain binds directly to a helical motif within the central intrinsically disordered region (IDR) of tensin3, whilst vinculin binds indirectly to tensin3 via talin. Using CRISPR knock-out cells in combination with defined tensin3 mutations, we show (i) that tensin3 is critical for the formation of α5β1-integrin FBs and for fibronectin fibrillogenesis, and (ii) the talin/tensin3 interaction drives this process, with vinculin acting to potentiate it.
Collapse
Affiliation(s)
- Paul Atherton
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK.,Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rafaella Konstantinou
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK.,sGSK Group, Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Suat Peng Neo
- Quantitative Proteomics Group, Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Emily Wang
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Eleonora Balloi
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Marina Ptushkina
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Hayley Bennett
- Genome Editing Unit, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Kath Clark
- Department of Biochemistry, University of Leicester, Leicester, UK
| | - Jayantha Gunaratne
- Quantitative Proteomics Group, Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - David Critchley
- Department of Biochemistry, University of Leicester, Leicester, UK
| | - Igor Barsukov
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Edward Manser
- sGSK Group, Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| |
Collapse
|
14
|
Zeng Y, Su X, Takezawa MG, Fichtinger PS, Lee UN, Pippin JW, Shankland SJ, Lim FY, Denlinger LC, Jarjour NN, Mathur SK, Sandbo N, Berthier E, Esnault S, Bernau K, Theberge AB. An open microfluidic coculture model of fibroblasts and eosinophils to investigate mechanisms of airway inflammation. Front Bioeng Biotechnol 2022; 10:993872. [PMID: 36246374 PMCID: PMC9558094 DOI: 10.3389/fbioe.2022.993872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/05/2022] [Indexed: 11/22/2022] Open
Abstract
Interactions between fibroblasts and immune cells play an important role in tissue inflammation. Previous studies have found that eosinophils activated with interleukin-3 (IL-3) degranulate on aggregated immunoglobulin G (IgG) and release mediators that activate fibroblasts in the lung. However, these studies were done with eosinophil-conditioned media that have the capacity to investigate only one-way signaling from eosinophils to fibroblasts. Here, we demonstrate a coculture model of primary normal human lung fibroblasts (HLFs) and human blood eosinophils from patients with allergy and asthma using an open microfluidic coculture device. In our device, the two types of cells can communicate via two-way soluble factor signaling in the shared media while being physically separated by a half wall. Initially, we assessed the level of eosinophil degranulation by their release of eosinophil-derived neurotoxin (EDN). Next, we analyzed the inflammation-associated genes and soluble factors using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and multiplex immunoassays, respectively. Our results suggest an induction of a proinflammatory fibroblast phenotype of HLFs following the coculture with degranulating eosinophils, validating our previous findings. Additionally, we present a new result that indicate potential impacts of activated HLFs back on eosinophils. This open microfluidic coculture platform provides unique opportunities to investigate the intercellular signaling between the two cell types and their roles in airway inflammation and remodeling.
Collapse
Affiliation(s)
- Yuting Zeng
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Xiaojing Su
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Meg G. Takezawa
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Paul S. Fichtinger
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Ulri N. Lee
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Jeffery W. Pippin
- Division of Nephrology, School of Medicine, University of Washington, Seattle, WA, United States
| | - Stuart J. Shankland
- Division of Nephrology, School of Medicine, University of Washington, Seattle, WA, United States
| | - Fang Yun Lim
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Loren C. Denlinger
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Nizar N. Jarjour
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Sameer K. Mathur
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Nathan Sandbo
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Erwin Berthier
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Stephane Esnault
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Ksenija Bernau
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Ashleigh B. Theberge
- Department of Chemistry, University of Washington, Seattle, WA, United States
- Department of Urology, School of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
15
|
BMP3 inhibits TGFβ2-mediated myofibroblast differentiation during wound healing of the embryonic cornea. NPJ Regen Med 2022; 7:36. [PMID: 35879352 PMCID: PMC9314337 DOI: 10.1038/s41536-022-00232-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/06/2022] [Indexed: 11/29/2022] Open
Abstract
Often acute damage to the cornea initiates drastic tissue remodeling, resulting in fibrotic scarring that disrupts light transmission and precedes vision impairment. Very little is known about the factors that can mitigate fibrosis and promote scar-free cornea wound healing. We previously described transient myofibroblast differentiation during non-fibrotic repair in an embryonic cornea injury model. Here, we sought to elucidate the mechanistic regulation of myofibroblast differentiation during embryonic cornea wound healing. We found that alpha-smooth muscle actin (αSMA)-positive myofibroblasts are superficial and their presence inversely correlates with wound closure. Expression of TGFβ2 and nuclear localization of pSMAD2 were elevated during myofibroblast induction. BMP3 and BMP7 were localized in the corneal epithelium and corresponded with pSMAD1/5/8 activation and absence of myofibroblasts in the healing stroma. In vitro analyses with corneal fibroblasts revealed that BMP3 inhibits the persistence of TGFβ2-induced myofibroblasts by promoting disassembly of focal adhesions and αSMA fibers. This was confirmed by the expression of vinculin and pFAK. Together, these data highlight a mechanism to inhibit myofibroblast persistence during cornea wound repair.
Collapse
|
16
|
Begum F, Keni R, Ahuja TN, Beegum F, Nandakumar K, Shenoy RR. Notch signaling: A possible therapeutic target and its role in diabetic foot ulcers. Diabetes Metab Syndr 2022; 16:102542. [PMID: 35724488 DOI: 10.1016/j.dsx.2022.102542] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND & AIM Diabetic foot ulcers are major cause of lower limb amputations in the diabetic population. The major factors that play a role in causing the delay of the process of healing in diabetic foot ulcers broadly are decreased angiogenesis, reduced proliferation and migration of keratinocytes/fibroblasts. The typical wound healing process has four phases which are overlapping with each other thus making the healing even more complex. Hence it is essential to identify a therapeutic target that involves the regulation of the cellular factors involved in healing and helps to increase angiogenesis and can regulate all four phases accordingly. METHOD Literature review involved a search of the databases namely, PubMed, Cochrane, EMBASE, and Web of Science database. Articles were identified and retrieved that specifically dealt with Notch as a target in healing of wounds and its mechanism of action on various cells and phases of healing. RESULTS Notch is a cell surface receptor which interacts with transmembrane ligands of the nearby cells and is involved in cell proliferation, differentiation, cell fate and death. It is also involved in cell-to-cell communication, cell signaling, and various phases of development. There exist four known notch genes and five ligands which interact with notch proteins. Hyperglycemia plays a role in the activation of the notch receptor thus causing the release of inflammatory mediators via macrophages. As notch can regulate macrophage-mediated inflammation it can serve as a therapeutic target for diabetic foot ulcers. CONCLUSION This review focuses on the effect of notch on various cell mediators and phases of diabetic wound healing and deals with how notch activation or inhibition can serve as a potential therapeutic target for healing diabetic foot ulcers.
Collapse
Affiliation(s)
- Farmiza Begum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Raghuvir Keni
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Tejas N Ahuja
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Fathima Beegum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Rekha R Shenoy
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
17
|
Bernau K, Skibba M, Leet JP, Furey S, Gehl C, Li Y, Zhou J, Sandbo N, Brasier AR. Selective Inhibition of Bromodomain-Containing Protein 4 Reduces Myofibroblast Transdifferentiation and Pulmonary Fibrosis. FRONTIERS IN MOLECULAR MEDICINE 2022; 2. [PMID: 35782526 PMCID: PMC9245900 DOI: 10.3389/fmmed.2022.842558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Idiopathic pulmonary fibrosis is a lethal disease driven by myofibroblast expansion. Currently no therapies exist that target the epigenetic mechanisms controlling myofibroblast transdifferentiation, which is responsible for unregulated extracellular matrix (ECM) production. We have recently shown that bromodomain-containing protein 4 (BRD4), an epigenetic regulator that forms a scaffold for nuclear activators and transcription factors, is essential for TGFβ-induced myofibroblast transdifferentiation. However, its role in the development and progression of pulmonary fibrosis in vivo has not been established. Here, we evaluate the hypothesis that BRD4 bromodomain interactions mediate myofibroblast expansion and fibrosing disease in vivo. C57BL/6J mice challenged with intratracheal bleomycin were systemically treated with a selective allosteric inhibitor of the BRD4 bromodomain 1 (BD1), ZL0591 (10 mg/kg), during the established fibrotic phase (14 days post-bleomycin) in a rigorous therapeutic paradigm. Eleven days after initiation of ZL0591 treatment (25 days post-bleomycin), we detected a significant improvement in blood O2 saturation compared to bleomycin/vehicle control. Twenty-eight days post-bleomycin, we observed a reduction in the volumetric Hounsfield Unit (HU) density by micro computed tomography (µCT) in the ZL0591-treated group, as well as a reduction in collagen deposition (hydroxyproline content) and severity of injury (Ashcroft scoring). Myofibroblast transdifferentiation was measured by smooth muscle α-actin (αSMA) staining, indicating a loss of this cell population in the ZL0591-treated group, and corresponded to reduced transcript levels of myofibroblast-associated extracellular matrix genes, tenascin-C and collagen 1α1. We conclude that BRD4 BD1 interactions are critical for myofibroblast transdifferentiation and fibrotic progression in a mouse model of pulmonary fibrosis.
Collapse
Affiliation(s)
- Ksenija Bernau
- Department of Medicine, School of Medicine and Public Health (SMPH), University of Wisconsin-Madison, Madison, WI, United States
- Correspondence: Ksenija Bernau,
| | - Melissa Skibba
- Department of Medicine, School of Medicine and Public Health (SMPH), University of Wisconsin-Madison, Madison, WI, United States
| | - Jonathan P. Leet
- Department of Medicine, School of Medicine and Public Health (SMPH), University of Wisconsin-Madison, Madison, WI, United States
| | - Sierra Furey
- Department of Medicine, School of Medicine and Public Health (SMPH), University of Wisconsin-Madison, Madison, WI, United States
| | - Carson Gehl
- Department of Medicine, School of Medicine and Public Health (SMPH), University of Wisconsin-Madison, Madison, WI, United States
| | - Yi Li
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Nathan Sandbo
- Department of Medicine, School of Medicine and Public Health (SMPH), University of Wisconsin-Madison, Madison, WI, United States
| | - Allan R. Brasier
- Department of Medicine, School of Medicine and Public Health (SMPH), University of Wisconsin-Madison, Madison, WI, United States
- Institute for Clinical and Translational Research (ICTR), University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
18
|
Spatiotemporal control of myofibroblast activation in acoustically-responsive scaffolds via ultrasound-induced matrix stiffening. Acta Biomater 2022; 138:133-143. [PMID: 34808418 PMCID: PMC8738148 DOI: 10.1016/j.actbio.2021.11.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 01/17/2023]
Abstract
Hydrogels are often used to study the impact of biomechanical and topographical cues on cell behavior. Conventional hydrogels are designed a priori, with characteristics that cannot be dynamically changed in an externally controlled, user-defined manner. We developed a composite hydrogel, termed an acoustically-responsive scaffold (ARS), that enables non-invasive, spatiotemporally controlled modulation of mechanical and morphological properties using focused ultrasound. An ARS consists of a phase-shift emulsion distributed in a fibrin matrix. Ultrasound non-thermally vaporizes the emulsion into bubbles, which induces localized, radial compaction and stiffening of the fibrin matrix. In this in vitro study, we investigate how this mechanism can control the differentiation of fibroblasts into myofibroblasts, a transition correlated with substrate stiffness on 2D substrates. Matrix compaction and stiffening was shown to be highly localized using confocal and atomic force microscopies, respectively. Myofibroblast phenotype, evaluated by α-smooth muscle actin (α-SMA) immunocytochemistry, significantly increased in matrix regions proximal to bubbles compared to distal regions, irrespective of the addition of exogenous transforming growth factor-β1 (TGF-β1). Introduction of the TGF-β1 receptor inhibitor SB431542 abrogated the proximal enhancement. This approach providing spatiotemporal control over biophysical signals and resulting cell behavior could aid in better understanding fibrotic disease progression and the development of therapeutic interventions for chronic wounds. STATEMENT OF SIGNIFICANCE: Hydrogels are used in cell culture to recapitulate both biochemical and biophysical aspects of the native extracellular matrix. Biophysical cues like stiffness can impact cell behavior. However, with conventional hydrogels, there is a limited ability to actively modulate stiffness after polymerization. We have developed an ultrasound-based method of spatiotemporally-controlling mechanical and morphological properties within a composite hydrogel, termed an acoustically-responsive scaffold (ARS). Upon exposure to ultrasound, bubbles are non-thermally generated within the fibrin matrix of an ARS, thereby locally compacting and stiffening the matrix. We demonstrate how ARSs control the differentiation of fibroblasts into myofibroblasts in 2D. This approach could assist with the study of fibrosis and the development of therapies for chronic wounds.
Collapse
|
19
|
Hypoxia induces stress fiber formation in adipocytes in the early stage of obesity. Sci Rep 2021; 11:21473. [PMID: 34728615 PMCID: PMC8563745 DOI: 10.1038/s41598-021-00335-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 10/07/2021] [Indexed: 02/07/2023] Open
Abstract
In obese adipose tissue (AT), hypertrophic expansion of adipocytes is not matched by new vessel formation, leading to AT hypoxia. As a result, hypoxia inducible factor-1⍺ (HIF-1⍺) accumulates in adipocytes inducing a transcriptional program that upregulates profibrotic genes and biosynthetic enzymes such as lysyl oxidase (LOX) synthesis. This excess synthesis and crosslinking of extracellular matrix (ECM) components cause AT fibrosis. Although fibrosis is a hallmark of obese AT, the role of fibroblasts, cells known to regulate fibrosis in other fibrosis-prone tissues, is not well studied. Here we have developed an in vitro model of AT to study adipocyte-fibroblast crosstalk in a hypoxic environment. Further, this in vitro model was used to investigate the effect of hypoxia on adipocyte mechanical properties via ras homolog gene family member A (RhoA)/Rho-associated coiled-coil kinases (ROCK) signaling pathways. We confirmed that hypoxia creates a diseased phenotype by inhibiting adipocyte maturation and inducing actin stress fiber formation facilitated by myocardin-related transcription factor A (MRTF-A/MKL1) nuclear translocation. This work presents new potential therapeutic targets for obesity by improving adipocyte maturation and limiting mechanical stress in obese AT.
Collapse
|
20
|
Luo C, Huang B, Wu Y, Xu Y, Ou W, Chen J, Chen L. Identification of Lymph Node Metastasis-Related Key Genes and Prognostic Risk Model in Bladder Cancer by Co-Expression Analysis. Front Mol Biosci 2021; 8:633299. [PMID: 34368222 PMCID: PMC8339436 DOI: 10.3389/fmolb.2021.633299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Lymph node metastasis (LNM) is an important pathological characteristic of bladder cancer (BCa). However, the molecular mechanism underlying LNM was not thoroughly elaborated. Identification for LNM-related biomarkers may contribute to making suitable therapies. So, the current study was aimed to identify key genes and construct a prognostic signature. Methods: Based on the Cancer Genome Atlas (TCGA) database, gene expression and clinical information were obtained. Then, the weighted gene co-expression network analysis (WGCNA) was performed to identify the key modules and hub genes. A function analysis and a gene set enrichment analysis were applied to explore biological functions and pathways of interested genes. Furthermore, a prognostic model based on LNM-related genes was constructed by using the least absolute shrinkage and selection operator (LASSO) Cox regression analysis. Results: Finally, nine co-expression modules were constructed, and two modules (turquoise and green) were significantly associated with LNM. Three hub genes were identified as DACT3, TNS1, and MSRB3, which were annotated in actin binding, actin cytoskeleton, adaptive immune response, and cell adhesion molecular binding by the GSEA method. Further analysis demonstrated that three hub genes were associated with the overall survival of BCa patients. In addition, we built a prognostic signature based on the genes from LNM-related modules and evaluated the prognostic value of this signature. Conclusion: In general, this study revealed the key genes related to LNM and prognostic signature, which might provide new insights into therapeutic target of BCa.
Collapse
Affiliation(s)
- Cheng Luo
- Department of Urology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Bin Huang
- Department of Urology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yukun Wu
- Department of Urology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yadong Xu
- Department of Urology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wei Ou
- Department of Urology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Junxing Chen
- Department of Urology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lingwu Chen
- Department of Urology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
21
|
Hannan RT, Miller AE, Hung RC, Sano C, Peirce SM, Barker TH. Extracellular matrix remodeling associated with bleomycin-induced lung injury supports pericyte-to-myofibroblast transition. Matrix Biol Plus 2021; 10:100056. [PMID: 34195593 PMCID: PMC8233458 DOI: 10.1016/j.mbplus.2020.100056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022] Open
Abstract
Of the many origins of pulmonary myofibroblasts, microvascular pericytes are a known source. Prior literature has established the ability of pericytes to transition into myofibroblasts, but provide limited insight into molecular cues that drive this process during lung injury repair and fibrosis. Fibronectin and RGD-binding integrins have long been considered pro-fibrotic factors in myofibroblast biology, and here we test the hypothesis that these known myofibroblast cues coordinate pericyte-to-myofibroblast transitions. Specifically, we hypothesized that αvβ3 integrin engagement on fibronectin induces pericyte transition into myofibroblastic phenotypes in the murine bleomycin lung injury model. Myosin Heavy Chain 11 (Myh11)-CreERT2 lineage tracing in transgenic mice allows identification of cells of pericyte origin and provides a robust tool for isolating pericytes from tissues for further evaluation. We used this murine model to track and characterize pericyte behaviors during tissue repair. The majority of Myh11 lineage-positive cells are positive for the pericyte surface markers, PDGFRβ (55%) and CD146 (69%), and display typical pericyte morphology with spatial apposition to microvascular networks. After intratracheal bleomycin treatment of mice, Myh11 lineage-positive cells showed significantly increased contractile and secretory markers, as well as αv integrin expression. According to RNASeq measurements, many disease and tissue-remodeling genesets were upregulated in Myh11 lineage-positive cells in response to bleomycin-induced lung injury. In vitro, blocking αvβ3 binding through cycloRGDfK prevented expression of the myofibroblastic marker αSMA relative to controls. In response to RGD-containing provisional matrix proteins present in lung injury, pericytes may alter their integrin profile.
Collapse
Affiliation(s)
- Riley T. Hannan
- Department of Pathology, University of Virginia, 415 Lane Road, Charlottesville, VA, United States
| | - Andrew E. Miller
- Department of Biomedical Engineering, University of Virginia, 415 Lane Road, Charlottesville, VA, United States
| | - Ruei-Chun Hung
- Department of Biomedical Engineering, University of Virginia, 415 Lane Road, Charlottesville, VA, United States
| | - Catherine Sano
- Department of Chemical Engineering, University of Virginia, 102 Engineer's Way, Charlottesville, VA, United States
| | - Shayn M. Peirce
- Department of Biomedical Engineering, University of Virginia, 415 Lane Road, Charlottesville, VA, United States
| | - Thomas H. Barker
- Department of Biomedical Engineering, University of Virginia, 415 Lane Road, Charlottesville, VA, United States
| |
Collapse
|
22
|
Wilkinson HN, Hardman MJ. A role for estrogen in skin ageing and dermal biomechanics. Mech Ageing Dev 2021; 197:111513. [PMID: 34044023 DOI: 10.1016/j.mad.2021.111513] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 01/11/2023]
Abstract
The skin is the body's primary defence against the external environment, preventing infection and desiccation. Therefore, alterations to skin homeostasis, for example with skin ageing, increase susceptibility to skin disease and injury. Skin biological ageing is uniquely influenced by a combination of intrinsic and extrinsic (primarily photoageing) factors, with differential effects on skin structure and function. Interestingly, skin architecture rapidly changes following the menopause, as a direct result of reduced circulating 17β-estradiol. The traditional clinical benefit of estrogens are supported by recent experimental data, where 17β-estradiol supplementation prevents age-related decline in the skin's structural and mechanical properties. However, the off-target effects of 17β-estradiol continue to challenge therapeutic application. Here we discuss how ageing alters the physiological and structural properties of the dermal extracellular matrix, and explore how estrogen receptor-targeted therapies may restore the mechanical defects associated with skin ageing.
Collapse
Affiliation(s)
- Holly N Wilkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, The University of Hull, HU6 7RX, United Kingdom
| | - Matthew J Hardman
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, The University of Hull, HU6 7RX, United Kingdom.
| |
Collapse
|
23
|
Spada S, Tocci A, Di Modugno F, Nisticò P. Fibronectin as a multiregulatory molecule crucial in tumor matrisome: from structural and functional features to clinical practice in oncology. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:102. [PMID: 33731188 PMCID: PMC7972229 DOI: 10.1186/s13046-021-01908-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022]
Abstract
Deciphering extracellular matrix (ECM) composition and architecture may represent a novel approach to identify diagnostic and therapeutic targets in cancer. Among the ECM components, fibronectin and its fibrillary assembly represent the scaffold to build up the entire ECM structure, deeply affecting its features. Herein we focus on this extraordinary protein starting from its complex structure and defining its role in cancer as prognostic and theranostic marker.
Collapse
Affiliation(s)
- Sheila Spada
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Annalisa Tocci
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Francesca Di Modugno
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.
| | - Paola Nisticò
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
24
|
Pseudosarcomatous myofibroblastic proliferations of the urinary bladder are neoplasms characterized by recurrent FN1-ALK fusions. Mod Pathol 2021; 34:469-477. [PMID: 32908253 DOI: 10.1038/s41379-020-00670-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 11/08/2022]
Abstract
Pseudosarcomatous myofibroblastic proliferation is a descriptive term that designates a group of clinically indolent genitourinary lesions that most commonly arise in the urinary bladder. Given that pseudosarcomatous myofibroblastic proliferation may show morphologic overlap with inflammatory myofibroblastic tumor, the relationship, if any, between the two entities has been unclear. Moreover, pseudosarcomatous myofibroblastic proliferations are known to be positive for ALK immunohistochemistry in a subset of cases, although an inconsistent association with ALK rearrangement (ranging from 0 to 60%) has been reported. The objectives of this study were to determine the frequency of ALK rearrangement and to identify fusion partners using fluorescence in situ hybridization (FISH) and targeted RNA sequencing studies in a contemporary series of 30 pseudosarcomatous myofibroblastic proliferations of the urinary bladder, as well as to investigate ROS1 status by immunohistochemistry. ALK immunohistochemistry was positive in 70% (21/30) of pseudosarcomatous myofibroblastic proliferations; ROS1 immunohistochemistry was consistently negative (0/28). ALK rearrangements were detected by FISH in 86% (18/21) of cases, correlating with ALK immunohistochemical positivity in all but 3 cases. Of eight cases confirmed to be ALK rearranged by FISH, targeted RNA-sequencing detected FN1-ALK fusions in seven (88%) cases, which involved exons 20-26 of FN1 (5') and exon 18-19 of ALK (3'). In conclusion, ALK rearrangements are frequent in pseudosarcomatous myofibroblastic proliferations, typically involving exon 19, and FN1 appears to be a consistent fusion partner. Given the significant clinicopathologic differences between inflammatory myofibroblastic tumor and pseudosarcomatous myofibroblastic proliferation, our findings provide further support for classification of pseudosarcomatous myofibroblastic proliferation as a distinct clinicopathologic entity, and propose the alternate terminology "pseudosarcomatous myofibroblastic neoplasm of the genitourinary tract."
Collapse
|
25
|
Duan J, Wang L, Shang L, Yang S, Wu H, Huang Y, Miao Y. miR-152/TNS1 axis inhibits non-small cell lung cancer progression through Akt/mTOR/RhoA pathway. Biosci Rep 2021; 41:BSR20201539. [PMID: 33269380 PMCID: PMC7785040 DOI: 10.1042/bsr20201539] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 12/30/2022] Open
Abstract
AIM The purpose of the present study was to explore the function and mechanism of tensin 1 (TNS1) in non-small cell lung cancer (NSCLC) progression. METHODS The expression of TNS1 in NSCLC cells and tissues was assessed by RT-PCR and Western blot. Besides, Kaplan-Meier survival analysis was recruited to explore the association between TNS1 and NSCLC. Cell growth was analyzed by MTT and flow cytometry assay, while cell metastasis was determined by wound healing and transwell assays. The targeting relationship between TNS1 and miR-152 was assessed by luciferase activity assays. And Western blot was employed to determine the expression of related proteins of Akt/mTOR/RhoA pathway. RESULTS TNS1 level was boosted in NSCLC cells and tissues, related to the prognosis of NSCLC patients. Furthermore, it was proved that TNS1 promoted the growth and metastasis of NSCLC cells via Akt/mTOR/RhoA pathway. And miR-152 targeted TNS1 to affect the progression of NSCLC. CONCLUSION miR-152/TNS1 axis inhibits the progression of NSCLC by Akt/mTOR/RhoA pathway.
Collapse
Affiliation(s)
- Jinjin Duan
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, P.R. China
| | - Li Wang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, P.R. China
| | - Liqun Shang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, P.R. China
| | - Shumei Yang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, P.R. China
| | - Hua Wu
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, P.R. China
| | - Yongcheng Huang
- Department of Pathology, Xi’an Central Hospital, Xi’an, Shaanxi 7100033, P.R. China
| | - Yi Miao
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, P.R. China
| |
Collapse
|
26
|
P KM, Sivashanmugam K, Kandasamy M, Subbiah R, Ravikumar V. Repurposing of histone deacetylase inhibitors: A promising strategy to combat pulmonary fibrosis promoted by TGF-β signalling in COVID-19 survivors. Life Sci 2020; 266:118883. [PMID: 33316266 PMCID: PMC7831549 DOI: 10.1016/j.lfs.2020.118883] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 01/18/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has rapidly spread around the world causing global public health emergency. In the last twenty years, we have witnessed several viral epidemics such as severe acute respiratory syndrome coronavirus (SARS-CoV), Influenza A virus subtype H1N1 and most recently Middle East respiratory syndrome coronavirus (MERS-CoV). There were tremendous efforts endeavoured globally by scientists to combat these viral diseases and now for SARS-CoV-2. Several drugs such as chloroquine, arbidol, remdesivir, favipiravir and dexamethasone are adopted for use against COVID-19 and currently clinical studies are underway to test their safety and efficacy for treating COVID-19 patients. As per World Health Organization reports, so far more than 16 million people are affected by COVID-19 with a recovery of close to 10 million and deaths at 600,000 globally. SARS-CoV-2 infection is reported to cause extensive pulmonary damages in affected people. Given the large number of recoveries, it is important to follow-up the recovered patients for apparent lung function abnormalities. In this review, we discuss our understanding about the development of long-term pulmonary abnormalities such as lung fibrosis observed in patients recovered from coronavirus infections (SARS-CoV and MERS-CoV) and probable epigenetic therapeutic strategy to prevent the development of similar pulmonary abnormalities in SARS-CoV-2 recovered patients. In this regard, we address the use of U.S. Food and Drug Administration (FDA) approved histone deacetylase (HDAC) inhibitors therapy to manage pulmonary fibrosis and their underlying molecular mechanisms in managing the pathologic processes in COVID-19 recovered patients.
Collapse
Affiliation(s)
- Krishna Murthy P
- Cancer Biology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | | | - Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India; Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi, India
| | - Rajasekaran Subbiah
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhauri, Madhya Pradesh, India
| | - Vilwanathan Ravikumar
- Cancer Biology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India.
| |
Collapse
|
27
|
Skibba M, Drelich A, Poellmann M, Hong S, Brasier AR. Nanoapproaches to Modifying Epigenetics of Epithelial Mesenchymal Transition for Treatment of Pulmonary Fibrosis. Front Pharmacol 2020; 11:607689. [PMID: 33384604 PMCID: PMC7770469 DOI: 10.3389/fphar.2020.607689] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a chronically progressive interstitial lung that affects over 3 M people worldwide and rising in incidence. With a median survival of 2-3 years, IPF is consequently associated with high morbidity, mortality, and healthcare burden. Although two antifibrotic therapies, pirfenidone and nintedanib, are approved for human use, these agents reduce the rate of decline of pulmonary function but are not curative and do not reverse established fibrosis. In this review, we discuss the prevailing epithelial injury hypothesis, wherein pathogenic airway epithelial cell-state changes known as Epithelial Mesenchymal Transition (EMT) promotes the expansion of myofibroblast populations. Myofibroblasts are principal components of extracellular matrix production that result in airspace loss and mortality. We review the epigenetic transition driving EMT, a process produced by changes in histone acetylation regulating mesenchymal gene expression programs. This mechanistic work has focused on the central role of bromodomain-containing protein 4 in mediating EMT and myofibroblast transition and initial preclinical work has provided evidence of efficacy. As nanomedicine presents a promising approach to enhancing the efficacy of such anti-IPF agents, we then focus on the state of nanomedicine formulations for inhalable delivery in the treatment of pulmonary diseases, including liposomes, polymeric nanoparticles (NPs), inorganic NPs, and exosomes. These nanoscale agents potentially provide unique properties to existing pulmonary therapeutics, including controlled release, reduced systemic toxicity, and combination delivery. NP-based approaches for pulmonary delivery thus offer substantial promise to modify epigenetic regulators of EMT and advance treatments for IPF.
Collapse
Affiliation(s)
- Melissa Skibba
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
| | - Adam Drelich
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, United States
| | - Michael Poellmann
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, United States
| | - Seungpyo Hong
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, United States
- Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
- Yonsei Frontier Lab, Department of Pharmacy, Yonsei University, Seoul, South Korea
| | - Allan R. Brasier
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
- Institute for Clinical and Translational Research (ICTR), University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
28
|
Teh N, Leow LJ. The Role of Actin in Muscle Spasms in a Case Series of Patients with Advanced Basal Cell Carcinoma Treated with a Hedgehog Pathway Inhibitor. Dermatol Ther (Heidelb) 2020; 11:293-299. [PMID: 33200373 PMCID: PMC7859007 DOI: 10.1007/s13555-020-00464-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Indexed: 11/29/2022] Open
Abstract
Basal cell carcinoma (BCC) is the most common cancer globally, with the incidence increasing worldwide by approximately 1% annually. While most cases of BCC can be treated surgically, advanced BCC often poses treatment challenges for patients unsuitable for, or refractory to, radiotherapy and surgery. Since the majority of cases of BCC demonstrate Hedgehog signaling hyperactivation, Hedgehog pathway inhibitors provide durable treatment options and improved clinical outcomes for patients with advanced BCC. One of the most common adverse events seen in patients taking Hedgehog inhibitors includes muscle spasms, which are hypothesized to occur because of calcium influx into the muscle cells. Here we present a case series of patients with muscle spasms during treatment with sonidegib and propose an alternate etiology related to increased actin expression.
Collapse
Affiliation(s)
- Natalie Teh
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia.
| | | |
Collapse
|
29
|
Shao H, Li Y, Pastar I, Xiao M, Prokupets R, Liu S, Yu K, Vazquez-Padron RI, Tomic-Canic M, Velazquez OC, Liu ZJ. Notch1 signaling determines the plasticity and function of fibroblasts in diabetic wounds. Life Sci Alliance 2020; 3:3/12/e202000769. [PMID: 33109684 PMCID: PMC7652398 DOI: 10.26508/lsa.202000769] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022] Open
Abstract
Fibroblasts play a pivotal role in wound healing. However, the molecular mechanisms determining the reparative response of fibroblasts remain unknown. Here, we identify Notch1 signaling as a molecular determinant controlling the plasticity and function of fibroblasts in modulating wound healing and angiogenesis. The Notch pathway is activated in fibroblasts of diabetic wounds but not in normal skin and non-diabetic wounds. Consistently, wound healing in the FSP-1 +/- ;ROSA LSL-N1IC+/+ mouse, in which Notch1 is activated in fibroblasts, is delayed. Increased Notch1 activity in fibroblasts suppressed their growth, migration, and differentiation into myofibroblasts. Accordingly, significantly fewer myofibroblasts and less collagen were present in granulation tissues of the FSP-1 +/- ;ROSA LSL-N1IC+/+ mice, demonstrating that high Notch1 activity inhibits fibroblast differentiation. High Notch1 activity in fibroblasts diminished their role in modulating the angiogenic response. We also identified that IL-6 is a functional Notch1 target and involved in regulating angiogenesis. These findings suggest that Notch1 signaling determines the plasticity and function of fibroblasts in wound healing and angiogenesis, unveiling intracellular Notch1 signaling in fibroblasts as potential target for therapeutic intervention in diabetic wound healing.
Collapse
Affiliation(s)
- Hongwei Shao
- Department of Surgery, Miller School of Medicine, University of Miami, Coral Gables, FL, USA
| | - Yan Li
- Department of Surgery, Miller School of Medicine, University of Miami, Coral Gables, FL, USA
| | - Irena Pastar
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, Miller School of Medicine, University of Miami, Coral Gables, FL, USA
| | - Min Xiao
- Department of Surgery, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rochelle Prokupets
- Department of Surgery, Miller School of Medicine, University of Miami, Coral Gables, FL, USA
| | - Sophia Liu
- Department of Surgery, Miller School of Medicine, University of Miami, Coral Gables, FL, USA
| | - Kerstin Yu
- Department of Surgery, Miller School of Medicine, University of Miami, Coral Gables, FL, USA
| | | | - Marjana Tomic-Canic
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, Miller School of Medicine, University of Miami, Coral Gables, FL, USA
| | - Omaida C Velazquez
- Department of Surgery, Miller School of Medicine, University of Miami, Coral Gables, FL, USA
| | - Zhao-Jun Liu
- Department of Surgery, Miller School of Medicine, University of Miami, Coral Gables, FL, USA
| |
Collapse
|
30
|
Ravikrishnan A, Zhang H, Fox JM, Jia X. Core-Shell Microfibers via Bioorthogonal Layer-by-Layer Assembly. ACS Macro Lett 2020; 9:1369-1375. [PMID: 35638624 DOI: 10.1021/acsmacrolett.0c00515] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A new technique is described for the construction of core-shell microfibers for biomedical applications. Fibrous scaffolds were fabricated by electrospinning, followed by covalent layer-by-layer deposition based on the rapid bioorthogonal reaction between s-tetrazines (Tz) and trans-cyclooctenes (TCOs). Electrospun poly(ε-caprolactone) (PCL) scaffolds were subjected to surface modifications to install tetrazine groups. The scaffolds were iteratively submerged in aqueous solutions of TCO-modified hyaluronic acid (HA-TCO) and tetrazine-modified hyaluronic acid (HA-Tz), resulting in the controlled growth of a cross-linked HA gel around individual microfibers. Integrin-binding motifs were covalently attached to the surface of the microfibers using TCO-conjugated RGD peptide. The scaffolds fostered the attachment and growth of primary porcine vocal fold fibroblasts without a significant induction of the myofibroblast phenotype. Stimulation with transforming growth factor beta (TGF-β) moderately enhanced fibroblast activation, and inhibition of the Rho/ROCK signaling pathway using Y27632 further decreased the expression of myofibroblastic markers. The bioorthogonally assembled scaffolds with a stiff PCL core and a soft HA shell may find application as therapeutic implants for the treatment of vocal fold scarring.
Collapse
Affiliation(s)
- Anitha Ravikrishnan
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - He Zhang
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Joseph M Fox
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, United States
| | - Xinqiao Jia
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| |
Collapse
|
31
|
Abstract
Fibronectin (FN) circulating in the blood and produced by cells provides the basis of the extracellular matrix (ECM) formed in healing acute wounds. The time-dependent deposition of FN by macrophages, its synthesis by fibroblasts and myofibroblasts, and later degradation in the remodeled granulation tissue are a prerequisite for successful healing of wounds. However, the pattern of FN expression and deposition in skin lesions is disturbed. The degradation of the ECM components including FN in varicose veins prevails over ECM synthesis and deposition. FN is inconspicuous in the fibrotic lesions in lipodermatosclerosis, while tenascin-C containing FN-like peptide sequences are prominent. FN is produced in large amounts by fibroblasts at the edge of venous ulcers but FN deposition at the wound bed is impaired. Both the proteolytic environment in the wounds and the changed function of the ulcer fibroblasts may be responsible for the poor healing of venous ulcers. The aim of this review is to describe the current knowledge of FN pathophysiology in chronic venous diseases. In view of the fact that FN plays a crucial role in organizing the ECM, further research focused on FN metabolism in venous diseases may bring results applicable to the treatment of the diseases.
Collapse
Affiliation(s)
- Jiri Kanta
- Faculty of Medicine, Charles University, Hradec Kralove, Czech Republic
| | - Anna Zavadakova
- Biomedical Center, Faculty of Medicine, Charles University, Pilsen, Czech Republic
| |
Collapse
|
32
|
Stahnke T, Gajda-Deryło B, Jünemann AG, Stachs O, Sterenczak KA, Rejdak R, Beck J, Schütz E, Möller S, Barrantes I, Warsow G, Struckmann S, Fuellen G. Suppression of the TGF-β pathway by a macrolide antibiotic decreases fibrotic responses by ocular fibroblasts in vitro. ROYAL SOCIETY OPEN SCIENCE 2020; 7:200441. [PMID: 33047019 PMCID: PMC7540802 DOI: 10.1098/rsos.200441] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/19/2020] [Indexed: 05/12/2023]
Abstract
To elucidate and to inhibit post-surgical fibrotic processes after trabeculectomy in glaucoma therapy, we measured gene expression in a fibrotic cell culture model, based on transforming growth factor TGF-β induction in primary human tenon fibroblasts (hTFs), and used Connectivity Map (CMap) data for drug repositioning. We found that specific molecular mechanisms behind fibrosis are the upregulation of actins, the downregulation of CD34, and the upregulation of inflammatory cytokines such as IL6, IL11 and BMP6. The macrolide antibiotic Josamycin (JM) reverses these molecular mechanisms according to data from the CMap, and we thus tested JM as an inhibitor of fibrosis. JM was first tested for its toxic effects on hTFs, where it showed no influence on cell viability, but inhibited hTF proliferation in a concentration-dependent manner. We then demonstrated that JM suppresses the synthesis of extracellular matrix (ECM) components. In hTFs stimulated with TGF-β1, JM specifically inhibited α-smooth muslce actin expression, suggesting that it inhibits the transformation of fibroblasts into fibrotic myofibroblasts. In addition, a decrease of components of the ECM such as fibronectin, which is involved in in vivo scarring, was observed. We conclude that JM may be a promising candidate for the treatment of fibrosis after glaucoma filtration surgery or drainage device implantation in vivo.
Collapse
Affiliation(s)
- Thomas Stahnke
- Department of Ophthalmology, Rostock University Medical Center, Rostock, Germany
| | - Beata Gajda-Deryło
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Anselm G. Jünemann
- Department of Ophthalmology, Rostock University Medical Center, Rostock, Germany
| | - Oliver Stachs
- Department of Ophthalmology, Rostock University Medical Center, Rostock, Germany
| | | | - Robert Rejdak
- Department of General Ophthalmology, Medical University in Lublin, Poland
| | - Julia Beck
- Chronix Biomedical GmbH, Göttingen, Germany
| | | | - Steffen Möller
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Israel Barrantes
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Gregor Warsow
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Stephan Struckmann
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
- SHIP-KEF, Institute for Community Medicine, Greifswald University Medical Center, Greifswald, Germany
- Authors for correspondence: Stephan Struckmann e-mail:
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
- Authors for correspondence: Georg Fuellen e-mail:
| |
Collapse
|
33
|
Li L, Wang G, Cheung A, Abdelhady W, Seidl K, Xiong YQ. MgrA Governs Adherence, Host Cell Interaction, and Virulence in a Murine Model of Bacteremia Due to Staphylococcus aureus. J Infect Dis 2020; 220:1019-1028. [PMID: 31177268 DOI: 10.1093/infdis/jiz219] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/26/2019] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND MgrA is an important global virulence gene regulator in Staphylococcus aureus. In the present study, the role of mgrA in host-pathogen interactions related to virulence was explored in both methicillin-resistant S. aureus (MRSA) and methicillin-susceptible S. aureus (MSSA) strains. METHODS In vitro susceptibilities to human defense peptides (HDPs), adherence to fibronectin (Fn) and endothelial cells (ECs), EC damage, α-toxin production, expression of global regulator (eg, agr RNAIII) and its downstream effectors (eg, α-toxin [hla] and Fn binding protein A [fnbA]), MgrA binding to fnbA promoter, and the effect on HDP-induced mprF and dltA expression were analyzed. The impact of mgrA on virulence was evaluated using a mouse bacteremia model. RESULTS mgrA mutants displayed significantly higher susceptibility to HDPs, which might be related to the decreased HDP-induced mprF and dltA expression but decreased Fn and EC adherence, EC damage, α-toxin production, agr RNAIII, hla and fnbA expression, and attenuated virulence in the bacteremia model as compared to their respective parental and mgrA-complemented strains. Importantly, direct binding of MgrA to the fnbA promoter was observed. CONCLUSIONS These results suggest that mgrA mediates host-pathogen interactions and virulence and may provide a novel therapeutic target for invasive S. aureus infections.
Collapse
Affiliation(s)
- Liang Li
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance
| | - Genzhu Wang
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance
| | | | - Wessam Abdelhady
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance
| | - Kati Seidl
- University Hospital of Zurich, Switzerland
| | - Yan Q Xiong
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance.,David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
34
|
Prazosin inhibits the proliferation and survival of acute myeloid leukaemia cells through down-regulating TNS1. Biomed Pharmacother 2020; 124:109731. [DOI: 10.1016/j.biopha.2019.109731] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023] Open
|
35
|
Słoniecka M, Danielson P. Acetylcholine decreases formation of myofibroblasts and excessive extracellular matrix production in an in vitro human corneal fibrosis model. J Cell Mol Med 2020; 24:4850-4862. [PMID: 32176460 PMCID: PMC7176861 DOI: 10.1111/jcmm.15168] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/11/2020] [Accepted: 02/15/2020] [Indexed: 02/06/2023] Open
Abstract
Acetylcholine (ACh) has been reported to play various physiological roles, including wound healing in the cornea. Here, we study the role of ACh in the transition of corneal fibroblasts into myofibroblasts, and in consequence its role in the onset of fibrosis, in an in vitro human corneal fibrosis model. Primary human keratocytes were obtained from healthy corneas. Vitamin C (VitC) and transforming growth factor‐β1 (TGF‐β1) were used to induce fibrosis in corneal fibroblasts. qRT‐PCR and ELISA analyses showed that gene expression and production of collagen I, collagen III, collagen V, lumican, fibronectin (FN) and alpha‐smooth muscle actin (α‐SMA) were reduced by ACh in quiescent keratocytes. ACh treatment furthermore decreased gene expression and production of collagen I, collagen III, collagen V, lumican, FN and α‐SMA during the transition of corneal fibroblasts into myofibroblasts, after induction of fibrotic process. ACh inhibited corneal fibroblasts from developing contractile activity during the process of fibrosis, as assessed with collagen gel contraction assay. Moreover, the effect of ACh was dependent on activation of muscarinic ACh receptors. These results show that ACh has an anti‐fibrotic effect in an in vitro human corneal fibrosis model, as it negatively affects the transition of corneal fibroblasts into myofibroblasts. Therefore, ACh might play a role in the onset of fibrosis in the corneal stroma.
Collapse
Affiliation(s)
- Marta Słoniecka
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Patrik Danielson
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden.,Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| |
Collapse
|
36
|
Tisler M, Alkmin S, Chang HY, Leet J, Bernau K, Sandbo N, Campagnola PJ. Analysis of fibroblast migration dynamics in idiopathic pulmonary fibrosis using image-based scaffolds of the lung extracellular matrix. Am J Physiol Lung Cell Mol Physiol 2020; 318:L276-L286. [PMID: 31774302 PMCID: PMC7052674 DOI: 10.1152/ajplung.00087.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by a profound remodeling of the collagen in the extracellular matrix (ECM), where the fibers become both denser and more highly aligned. However, it is unknown how this reconfiguration of the collagen matrix affects disease progression. Here, we investigate the role of specific alterations in collagen fiber organization on cell migration dynamics by using biomimetic image-based collagen scaffolds representing normal and fibrotic lung, where the designs are derived directly from high-resolution second harmonic generation microscopy images. The scaffolds are fabricated by multiphoton-excited (MPE) polymerization, where the process is akin to three-dimensional printing, except that it is performed at much greater resolution (∼0.5 microns) and with collagen and collagen analogs. These scaffolds were seeded with early passaged primary human normal and IPF fibroblasts to enable the decoupling of the effect of cell-intrinsic characteristics (normal vs. IPF) versus ECM structure (normal vs. IPF) on migration dynamics. We found that the highly aligned IPF collagen structure promoted enhanced cell elongation and F-actin alignment along with increased cell migration speed and straightness relative to the normal tissues. Collectively, the data are consistent with an enhanced contact guidance mechanism on the aligned IPF matrix. Although cell intrinsic effects were observed, the aligned collagen matrix morphology had a larger effect on these metrics. Importantly, these biomimetic models of the lung cannot be synthesized by conventional fabrication methods. We suggest that the MPE image-based fabrication method will enable additional hypothesis-based testing studies of cell-matrix interactions in the context of tissue fibrosis.
Collapse
Affiliation(s)
- Marisa Tisler
- 1Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Samuel Alkmin
- 1Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Hsin-Yu Chang
- 1Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jon Leet
- 2Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ksenija Bernau
- 2Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Nathan Sandbo
- 2Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Paul J. Campagnola
- 1Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
37
|
Filla MS, Faralli JA, Desikan H, Peotter JL, Wannow AC, Peters DM. Activation of αvβ3 Integrin Alters Fibronectin Fibril Formation in Human Trabecular Meshwork Cells in a ROCK-Independent Manner. Invest Ophthalmol Vis Sci 2020; 60:3897-3913. [PMID: 31529121 PMCID: PMC6750892 DOI: 10.1167/iovs.19-27171] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Purpose Fibronectin fibrillogenesis is an integrin-mediated process that may contribute to the pathogenesis of primary open-angle glaucoma (POAG). Here, we examined the effects of αvβ3 integrins on fibrillogenesis in immortalized TM-1 cells and human trabecular meshwork (HTM) cells. Methods TM-1 cells overexpressing wild-type β3 (WTβ3) or constitutively active β3 (CAβ3) integrin subunits were generated. Control cells were transduced with an empty vector (EV). Deoxycholic acid (DOC) extraction of monolayers, immunofluorescence microscopy, and On-cell western analyses were used to determine levels of fibronectin fibrillogenesis and fibronectin fibril composition (EDA+ and EDB+ fibronectins) and conformation. αvβ3 and α5β1 Integrin levels were determined using fluorescence-activated cell sorting (FACS). Cilengitide and an adenovirus vector expressing WTβ3 or CAβ3 integrin subunits were used to examine the role of αvβ3 integrin in HTM cells. The role of the canonical α5β1 integrin–mediated pathway in fibrillogenesis was determined using the fibronectin-binding peptide FUD, the β1 integrin function-blocking antibody 13, and the Rho kinase (ROCK) inhibitor Y27632. Results Activation of αvβ3 integrin enhanced the assembly of fibronectin into DOC-insoluble fibrils in both TM-1 and HTM cells. The formation of fibronectin fibrils was dependent on α5β1 integrin and could be inhibited by FUD. However, fibrillogenesis was unaffected by Y27632. Fibrils assembled by CAβ3 cells also contained high levels of EDA+ and EDB+ fibronectin and fibronectin that was stretched. Conclusions αvβ3 Integrin signaling altered the deposition and structure of fibronectin fibrils using a β1 integrin/ROCK-independent mechanism. Thus, αvβ3 integrins could play a significant role in altering the function of fibronectin matrices in POAG.
Collapse
Affiliation(s)
- Mark S Filla
- Departments of Pathology & Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Jennifer A Faralli
- Departments of Pathology & Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Harini Desikan
- Departments of Pathology & Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Jennifer L Peotter
- Departments of Pathology & Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Abigail C Wannow
- Departments of Pathology & Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Donna M Peters
- Departments of Pathology & Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States.,Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| |
Collapse
|
38
|
Hiepen C, Jatzlau J, Hildebrandt S, Kampfrath B, Goktas M, Murgai A, Cuellar Camacho JL, Haag R, Ruppert C, Sengle G, Cavalcanti-Adam EA, Blank KG, Knaus P. BMPR2 acts as a gatekeeper to protect endothelial cells from increased TGFβ responses and altered cell mechanics. PLoS Biol 2019; 17:e3000557. [PMID: 31826007 PMCID: PMC6927666 DOI: 10.1371/journal.pbio.3000557] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 12/23/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022] Open
Abstract
Balanced transforming growth factor-beta (TGFβ)/bone morphogenetic protein (BMP)-signaling is essential for tissue formation and homeostasis. While gain in TGFβ signaling is often found in diseases, the underlying cellular mechanisms remain poorly defined. Here we show that the receptor BMP type 2 (BMPR2) serves as a central gatekeeper of this balance, highlighted by its deregulation in diseases such as pulmonary arterial hypertension (PAH). We show that BMPR2 deficiency in endothelial cells (ECs) does not abolish pan-BMP-SMAD1/5 responses but instead favors the formation of mixed-heteromeric receptor complexes comprising BMPR1/TGFβR1/TGFβR2 that enable enhanced cellular responses toward TGFβ. These include canonical TGFβ-SMAD2/3 and lateral TGFβ-SMAD1/5 signaling as well as formation of mixed SMAD complexes. Moreover, BMPR2-deficient cells express genes indicative of altered biophysical properties, including up-regulation of extracellular matrix (ECM) proteins such as fibrillin-1 (FBN1) and of integrins. As such, we identified accumulation of ectopic FBN1 fibers remodeled with fibronectin (FN) in junctions of BMPR2-deficient ECs. Ectopic FBN1 deposits were also found in proximity to contractile intimal cells in pulmonary artery lesions of BMPR2-deficient heritable PAH (HPAH) patients. In BMPR2-deficient cells, we show that ectopic FBN1 is accompanied by active β1-integrin highly abundant in integrin-linked kinase (ILK) mechano-complexes at cell junctions. Increased integrin-dependent adhesion, spreading, and actomyosin-dependent contractility facilitates the retrieval of active TGFβ from its latent fibrillin-bound depots. We propose that loss of BMPR2 favors endothelial-to-mesenchymal transition (EndMT) allowing cells of myo-fibroblastic character to create a vicious feed-forward process leading to hyperactivated TGFβ signaling. In summary, our findings highlight a crucial role for BMPR2 as a gatekeeper of endothelial homeostasis protecting cells from increased TGFβ responses and integrin-mediated mechano-transduction.
Collapse
Affiliation(s)
- Christian Hiepen
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Jerome Jatzlau
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Germany
| | - Susanne Hildebrandt
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Germany
| | - Branka Kampfrath
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Melis Goktas
- Max Planck Institute of Colloids and Interfaces, Mechano(bio)chemistry, Potsdam, Germany
| | - Arunima Murgai
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | | | - Rainer Haag
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Clemens Ruppert
- Universities of Giessen and Marburg Lung Center (UGMLC), Medical Clinic II, Justus Liebig University, Giessen, Germany
| | - Gerhard Sengle
- University of Cologne, Center for Biochemistry, Medical Faculty, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
| | | | - Kerstin G. Blank
- Max Planck Institute of Colloids and Interfaces, Mechano(bio)chemistry, Potsdam, Germany
| | - Petra Knaus
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| |
Collapse
|
39
|
Reduced Iron in Diabetic Wounds: An Oxidative Stress-Dependent Role for STEAP3 in Extracellular Matrix Deposition and Remodeling. J Invest Dermatol 2019; 139:2368-2377.e7. [DOI: 10.1016/j.jid.2019.05.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/16/2019] [Accepted: 05/23/2019] [Indexed: 12/18/2022]
|
40
|
James DS, Jambor AN, Chang HY, Alden Z, Tilbury KB, Sandbo NK, Campagnola PJ. Probing ECM remodeling in idiopathic pulmonary fibrosis via second harmonic generation microscopy analysis of macro/supramolecular collagen structure. JOURNAL OF BIOMEDICAL OPTICS 2019; 25:1-13. [PMID: 31785093 PMCID: PMC7008503 DOI: 10.1117/1.jbo.25.1.014505] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/06/2019] [Indexed: 05/06/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease with poor prognosis with short lifespan following diagnosis as patients have limited effective treatment options. A fundamental limitation is a lack of knowledge of the underlying collagen alterations in the disease, as this could lead to better diagnostics, prognostics, and measures of treatment efficacy. While the fibroses is the primary presentation of the disease, the collagen architecture has not been well studied beyond standard histology. Here, we used several metrics based on second harmonic generation (SHG) microscopy and optical scattering measurements to characterize the subresolution collagen assembly in human IPF and normal lung tissues. Using SHG directional analysis, we found that while collagen synthesis is increased in IPF, the resulting average fibril architecture is more disordered than in normal tissue. Wavelength-dependent optical scattering measurements lead to the same conclusion, and both optical approaches are consistent with ultrastructural analysis. SHG circular dichroism revealed significant differences in the net chirality between the fibrotic and normal collagen, where the former has a more randomized helical structure. Collectively, the measurements reveal significant changes in the collagen macro/supramolecular structure in the abnormal fibrotic collagen, and we suggest these alterations can serve as biomarkers for IPF diagnosis and progression.
Collapse
Affiliation(s)
- Darian S. James
- University of Wisconsin–Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | - Alexander N. Jambor
- University of Wisconsin–Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | - Hsin-Yu Chang
- University of Wisconsin–Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | - Zachary Alden
- University of Wisconsin–Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | - Karissa B. Tilbury
- University of Wisconsin–Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | - Nathan K. Sandbo
- University of Wisconsin–Madison, Division of Allergy, Pulmonary, and Critical Care Medicine, Madison, Wisconsin, United States
| | - Paul J. Campagnola
- University of Wisconsin–Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
- Address all correspondence to Paul J. Campagnola, E-mail:
| |
Collapse
|
41
|
Chen G, Xia B, Fu Q, Huang X, Wang F, Chen Z, Lv Y. Matrix Mechanics as Regulatory Factors and Therapeutic Targets in Hepatic Fibrosis. Int J Biol Sci 2019; 15:2509-2521. [PMID: 31754325 PMCID: PMC6854372 DOI: 10.7150/ijbs.37500] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/07/2019] [Indexed: 12/12/2022] Open
Abstract
The hallmark of liver fibrosis is excessive extracellular matrix (ECM) synthesis and deposition that improve liver matrix remodeling and stiffening. Increased matrix stiffness is not only a pathological consequence of liver fibrosis in traditional view, but also recognized as a key driver in pathological progression of hepatic fibrosis. Cells can perceive changes in the mechanical characteristics of hepatic matrix and respond by means of mechanical signal transduction pathways to regulate cell behavior. In this review, the authors first classify causes of liver matrix stiffening during fibrotic progression, such as higher degree of collagen cross-linking. The latest advances of the research on the matrix mechanics in regulating activation of HSCs or fibroblasts under two-dimensional (2D) and three-dimensional (3D) microenvironment is also classified and summarized. The mechanical signaling pathways involved in the process of hepatic matrix stiffening, such as YAP-TAZ signaling pathway, are further summarized. Finally, some potential therapeutic concepts and strategies based on matrix mechanics will be detailed. Collectively, these findings reinforce the importance of matrix mechanics in hepatic fibrosis, and underscore the value of clarifying its modulation in hopes of advancing the development of novel therapeutic targets and strategies for hepatic fibrosis.
Collapse
Affiliation(s)
- Guobao Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China
| | - Bin Xia
- Chongqing Technology and Business University, Chongqing 400067, P. R. China
| | - Qiang Fu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China
| | - Xiang Huang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China
| | - Fuping Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China
| | - Zhongmin Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China
| | - Yonggang Lv
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, P. R. China
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
42
|
Zbyszynski P, Toraason I, Repp L, Kwon GS. Probing the subcutaneous absorption of a PEGylated FUD peptide nanomedicine via in vivo fluorescence imaging. NANO CONVERGENCE 2019; 6:22. [PMID: 31281949 PMCID: PMC6612524 DOI: 10.1186/s40580-019-0192-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/12/2019] [Indexed: 05/04/2023]
Abstract
The Functional Upstream Domain (FUD) peptide is a potent inhibitor of fibronectin assembly and a therapeutic candidate for disorders linked with hyperdeposition of fibronectin-modulated ECM proteins. Most recently, experiments involving subcutaneous (s.c.) administration of a PEGylated FUD (PEG-FUD) of 27.5 kDa molecular weight yielded a significant reduction of fibronectin and collagen deposition in a murine model of renal fibrosis. The benefits of FUD PEGylation need to be studied to unlock the full potential of the PEG-FUD platform. This work studies the impact of PEGylating the FUD peptide with differently sized PEG on its absorption from the site of injection following s.c. delivery using non-invasive in vivo fluorescence imaging. The FUD and mFUD (control) peptides and their 10 kDa, 20 kDa, and 40 kDa PEG conjugates were labeled with the sulfo-Cy5 fluorophore. Isothermal titration calorimetry (ITC) and confocal fluorescence microscopy experiments verified FUD and PEG-FUD fibronectin binding activity preservation following sulfo-Cy5 labeling. Fluorescence in vivo imaging experiments revealed a linear relationship between the absorption apparent half-life (t1/2) and the MW of FUD, mFUD, and their PEG conjugates. Detected drug signal in the kidney and bladder regions of mice suggests that smaller peptides of both the FUD and mFUD series enter the kidney earlier and in higher amounts than their larger PEG conjugates. This work highlights an important delayed dose absorption enhancement that MW modification via PEGylation can contribute to a drug when combined with the subcutaneous route of delivery.
Collapse
Affiliation(s)
- Pawel Zbyszynski
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Inger Toraason
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Lauren Repp
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Glen S Kwon
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
43
|
Zhang M, Urabe G, Little C, Wang B, Kent AM, Huang Y, Kent KC, Guo LW. HDAC6 Regulates the MRTF-A/SRF Axis and Vascular Smooth Muscle Cell Plasticity. JACC Basic Transl Sci 2018; 3:782-795. [PMID: 30623138 PMCID: PMC6314972 DOI: 10.1016/j.jacbts.2018.08.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 07/31/2018] [Accepted: 08/23/2018] [Indexed: 01/04/2023]
Abstract
Distinct from other histone deacetylases, HDAC6 primarily resides in the cytosol. Unexpectedly, HDAC6-selective inhibition (or silencing) enhances the nuclear activity of SRF. HDAC6 inhibition elevates acetylation and protein levels of myocardin-related transcription factor A, a cytoplasmic-nuclear shuttling co-activator of SRF. Myocardin-related transcription factor A/SRF are known to critically regulate vascular smooth muscle cell phenotypic stability. HDAC6 inhibition prevents smooth muscle cell dedifferentiation in vitro and reduces neointima and restenosis in vivo.
Cellular plasticity is fundamental in biology and disease. Vascular smooth muscle cell (SMC) dedifferentiation (loss of contractile proteins) initiates and perpetrates vascular pathologies such as restenosis. Contractile gene expression is governed by the master transcription factor, serum response factor (SRF). Unlike other histone deacetylases, histone deacetylase 6 (HDAC6) primarily resides in the cytosol. Whether HDAC6 regulates SRF nuclear activity was previously unknown in any cell type. This study found that selective inhibition of HDAC6 with tubastatin A preserved the contractile protein (alpha-smooth muscle actin) that was otherwise diminished by platelet-derived growth factor-BB. Tubastatin A also enhanced SRF transcriptional (luciferase) activity, and this effect was confirmed by HDAC6 knockdown. Interestingly, HDAC6 inhibition increased acetylation and total protein of myocardin-related transcription factor A (MRTF-A), a transcription co-activator known to translocate from the cytosol to the nucleus, thereby activating SRF. Consistently, HDAC6 co-immunoprecipitated with MRTF-A. In vivo studies showed that tubastatin A treatment of injured rat carotid arteries mitigated neointimal lesion, which is known to be formed largely by dedifferentiated SMCs. This report is the first to show HDAC6 regulation of the MRTF-A/SRF axis and SMC plasticity, thus opening a new perspective for interventions of vascular pathologies.
Collapse
Key Words
- DMEM, Dulbecco’s modified Eagle’s medium
- DNA, deoxyribonucleic acid
- EEL, external elastic lamina
- FBS, fetal bovine serum
- HDAC, histone deacetylase
- HDAC6
- IEL, internal elastic lamina
- IH, intimal hyperplasia
- IgG, immunoglobulin G
- MMP, matrix metalloproteinase
- MRTF-A
- MRTF-A, myocardin-related transcription factor A
- PDGF-BB, platelet-derived growth factor-BB
- SMA, smooth muscle actin
- SMC, vascular smooth muscle cell
- SMHC, smooth muscle myosin heavy chain
- SRF
- SRF, serum response factor
- TNF, tumor necrosis factor
- TSA, trichostatin A
- dedifferentiation
- siRNA, small interfering ribonucleic acid
- vascular smooth muscle cell
Collapse
Affiliation(s)
- Mengxue Zhang
- Department of Surgery and Department of Physiology and Cell Biology, College of Medicine, and the Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, Ohio.,Cellular and Molecular Pathology Graduate Program, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Go Urabe
- Department of Surgery and Department of Physiology and Cell Biology, College of Medicine, and the Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, Ohio.,Department of Surgery, College of Medicine, and the Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Christopher Little
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Bowen Wang
- Department of Surgery, College of Medicine, and the Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Alycia M Kent
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Yitao Huang
- Department of Surgery and Department of Physiology and Cell Biology, College of Medicine, and the Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - K Craig Kent
- Department of Surgery, College of Medicine, and the Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Lian-Wang Guo
- Department of Surgery and Department of Physiology and Cell Biology, College of Medicine, and the Davis Heart and Lung Research Institute, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
44
|
PEGylated pUR4/FUD peptide inhibitor of fibronectin fibrillogenesis decreases fibrosis in murine Unilateral Ureteral Obstruction model of kidney disease. PLoS One 2018; 13:e0205360. [PMID: 30356276 PMCID: PMC6200241 DOI: 10.1371/journal.pone.0205360] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 09/24/2018] [Indexed: 01/06/2023] Open
Abstract
Fibronectin is a blood and extracellular matrix glycoprotein that plays important roles in wound healing and fibrosis since it controls the deposition of collagen and other extracellular matrix molecules and is a substrate for infiltrating lymphocytes. Using a high-affinity fibronectin-binding peptide (FUD/pUR4) that inhibits fibronectin deposition into extracellular matrix (ECM), we tested the ability of a PEGylated FUD/pUR4 (PEG-FUD) to inhibit fibrosis in the Unilateral Ureteral Obstruction (UUO) kidney disease model. Fibronectin fibrillogenesis assays, using human fibroblasts and human proximal tubular epithelial cultures, showed that PEG-FUD can inhibit fibronectin fibrillogenesis in vitro with an IC50 similar to unconjugated FUD, in the order of 20–35 nM. In contrast, a mutated FUD (mFUD) conjugated to PEG that lacked activity did not inhibit fibronectin assembly, even at 20 μM. The in vivo activity of PEG-FUD was tested in the murine UUO model by daily subcutaneous injection of 12.5 mg/kg for 7 days until harvest at day 10. Control treatments included saline, PEG, unconjugated FUD, and PEG-mFUD. Immunoblotting studies showed that fibronectin was enriched in the extracellular matrix fractions of extracted UUO kidneys, compared to contralateral untreated kidneys. In vivo, PEG-FUD significantly decreased fibronectin by ~70% in UUO kidneys as determined by both IHC and immunoblotting, respectively. In contrast, neither PEG-mFUD, PEG, nor saline had any significant effect. PEG-FUD also decreased collagens I and III and CD45-expressing cells (leukocytes) by ~60% and ~50%, as ascertained by picrosirius red staining and IHC, respectively. Immunoblotting studies also showed that the fibronectin remaining after PEG-FUD treatment was intact. Utilizing a custom-made polyclonal antibody generated against pUR4/FUD, intact PEG-FUD was detected by immunoblotting in both the ECM and lysate fractions of UUO kidneys. No adverse reaction or event was noted with any treatment. In summary, these studies suggest that PEG-FUD reached the kidneys without degradation, and decreased fibronectin incorporation into interstitial tissue. Decreased fibronectin was accompanied by a decrease in collagen and leukocyte infiltration. We propose that PEG-FUD, a specific inhibitor of fibronectin assembly, may be a candidate therapeutic for the treatment of fibrosis in kidney diseases.
Collapse
|
45
|
Zahir-Jouzdani F, Khonsari F, Soleimani M, Mahbod M, Arefian E, Heydari M, Shahhosseini S, Dinarvand R, Atyabi F. Nanostructured lipid carriers containing rapamycin for prevention of corneal fibroblasts proliferation and haze propagation after burn injuries: In vitro and in vivo. J Cell Physiol 2018; 234:4702-4712. [PMID: 30191977 DOI: 10.1002/jcp.27243] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/25/2018] [Indexed: 01/25/2023]
Abstract
Chemical burns are a major cause of corneal haze and blindness. Corticosteroids are commonly used after corneal burns to attenuate the severity of the inflammation-related fibrosis. While research efforts have been aimed toward application of novel therapeutics. In the current study, a novel drug delivery system based nanostructured lipid carriers (NLCs) were designed to treat corneal alkaline burn injury. Rapamycin, a potent inhibitor of mammalian target of rapamycin pathway, was loaded in NLCs (rapa-NLCs), and the NLCs were characterized. Cell viability assay, cellular uptake of NLCs, and in vitro evaluation of the fibrotic/angiogenic genes suppression by rapa-NLCs were carried out on human isolated corneal fibroblasts. Immunohistochemistry (IHC) assays were also performed after treatment of murine model of corneal alkaline burn with rapa-NLCs. According to the results, rapamycin was efficiently loaded in NLCs. NLCs could enhance coumarin-6 fibroblast uptake by 1.5 times. Rapa-NLCs efficiently downregulated platelet-derived growth factor and transforming growth factor beta genes in vitro. Furthermore, proliferation of fibroblasts, a major cause of corneal haze after injury, reduced. IHC staining of treated cornea with alpha-smooth muscle actin and CD34 + antibodies showed efficient prevention of myofibroblasts differentiation and angiogenesis, respectively. In conclusion, ocular delivery of rapamycin using NLCs after corneal injury may be considered as a promising antifibrotic/angiogenic treatment approach to preserve patient eyesight.
Collapse
Affiliation(s)
- Forouhe Zahir-Jouzdani
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Khonsari
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Bonyakhteh Stem Cell Research Center, Cellular and Molecular Biology Department, Tehran, Iran
| | - Mirgholamreza Mahbod
- Noor Ophthalmology Research Center, Pathology Department, Noor Eye Hospital, Tehran, Iran
| | - Ehsan Arefian
- Bonyakhteh Stem Cell Research Center, Cellular and Molecular Biology Department, Tehran, Iran.,Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Mostafa Heydari
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Shahhosseini
- Noor Ophthalmology Research Center, Pathology Department, Noor Eye Hospital, Tehran, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Nanotechnology Research Centre, Novel Drug Delivery Department, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Nanotechnology Research Centre, Novel Drug Delivery Department, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Roman W, Martins JP, Gomes ER. Local Arrangement of Fibronectin by Myofibroblasts Governs Peripheral Nuclear Positioning in Muscle Cells. Dev Cell 2018; 46:102-111.e6. [PMID: 29937388 PMCID: PMC6035285 DOI: 10.1016/j.devcel.2018.05.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 04/20/2018] [Accepted: 05/25/2018] [Indexed: 12/24/2022]
Abstract
Skeletal muscle cells (myofibers) are rod-shaped multinucleated cells surrounded by an extracellular matrix (ECM) basal lamina. In contrast to other cell types, nuclei in myofibers are positioned just below the plasma membrane at the cell periphery. Peripheral nuclear positioning occurs during myogenesis and is driven by myofibril crosslinking and contraction. Here we show that peripheral nuclear positioning is triggered by local accumulation of fibronectin secreted by myofibroblasts. We demonstrate that fibronectin via α5-integrin mediates peripheral nuclear positioning dependent on FAK and Src activation. Finally, we show that Cdc42, downstream of restricted fibronectin activation, is required for myofibril crosslinking but not myofibril contraction. Thus we identify that local activation of integrin by fibronectin secreted by myofibroblasts activates peripheral nuclear positioning in skeletal myofibers.
Collapse
Affiliation(s)
- William Roman
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal.
| | - João P Martins
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Edgar R Gomes
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal.
| |
Collapse
|
47
|
Bell R, Gendron NR, Anderson M, Flatow EL, Andarawis-Puri N. A potential new role for myofibroblasts in remodeling of sub-rupture fatigue tendon injuries by exercise. Sci Rep 2018; 8:8933. [PMID: 29895865 PMCID: PMC5997675 DOI: 10.1038/s41598-018-27196-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/30/2018] [Indexed: 12/14/2022] Open
Abstract
Tendons are ineffective at repairing sub-rupture fatigue injuries. Accordingly, we evaluated whether an exercise protocol that we have previously found to decrease structural damage kinks in fatigue damaged tendons, leads to improvement in mechanical properties. We hypothesized that exercise that promotes repair of fatigue damage will decrease apoptosis and increase the population of myofibroblasts. Rat patellar tendons underwent in vivo fatigue loading for 500 or 7200 cycles. Animals resumed cage activity for 2-weeks, then either remained cage active or began treadmill running until sacrifice at 4- or 10-weeks post-fatigue loading. Exercise following fatigue damage increased the stiffness back towards naïve levels, decreased apoptosis and increased the population of myofibroblasts. Next, proteins associated with inhibition of apoptosis (Collagen VI) or activation of myofibroblast (pSmad 2/3, fibrillin, integrin subunits αV and α5) were evaluated. Data suggests that collagen VI may not be integral to inhibition of apoptosis in this context. Exercise increased pSmad 2/3 and fibrillin in the insertion region for the 7200-cycles group. In addition, exercise decreased integrin αV and increased integrin α5 in fatigue damaged tendons. Data suggests that a decrease in apoptosis and an increase in population of myofibroblasts may be integral to remodeling of fatigue damaged tendons.
Collapse
Affiliation(s)
- Rebecca Bell
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | - N Remi Gendron
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew Anderson
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evan L Flatow
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nelly Andarawis-Puri
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA. .,Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA. .,Hospital for Special Surgery, New York, NY, USA.
| |
Collapse
|
48
|
Noskovičová N, Heinzelmann K, Burgstaller G, Behr J, Eickelberg O. Cub domain-containing protein 1 negatively regulates TGF-β signaling and myofibroblast differentiation. Am J Physiol Lung Cell Mol Physiol 2018; 314:L695-L707. [PMID: 29351434 DOI: 10.1152/ajplung.00205.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Fibroblasts are thought to be the prime cell type for producing and secreting extracellular matrix (ECM) proteins in the connective tissue. The profibrotic cytokine transforming growth factor-β1 (TGF-β1) activates and transdifferentiates fibroblasts into α-smooth muscle actin (α-SMA)-expressing myofibroblasts, which exhibit increased ECM secretion, in particular collagens. Little information, however, exists about cell-surface molecules on fibroblasts that mediate this transdifferentiation process. We recently identified, using unbiased cell-surface proteome analysis, Cub domain-containing protein 1 (CDCP1) to be strongly downregulated by TGF-β1. CDCP1 is a transmembrane glycoprotein, the expression and role of which has not been investigated in lung fibroblasts to date. Here, we characterized, in detail, the effect of TGF-β1 on CDCP1 expression and function, using immunofluorescence, FACS, immunoblotting, and siRNA-mediated knockdown of CDCP1. CDCP1 is present on interstitial fibroblasts, but not myofibroblasts, in the normal and idiopathic pulmonary fibrosis lung. In vitro, TGF-β1 decreased CDCP1 expression in a time-dependent manner by impacting mRNA and protein levels. Knockdown of CDCP1 enhanced a TGF-β1-mediated cell adhesion of fibroblasts. Importantly, CDCP1-depleted cells displayed an enhanced expression of profibrotic markers, such as collagen V or α-SMA, which was found to be independent of TGF-β1. Our data show, for the very first time that loss of CDCP1 contributes to fibroblast to myofibroblast differentiation via a potential negative feedback loop between CDCP1 expression and TGF-β1 stimulation.
Collapse
Affiliation(s)
- Nina Noskovičová
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the CPC-M BioArchive, Member of the German Center for Lung Research (DZL) , Munich , Germany
| | - Katharina Heinzelmann
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the CPC-M BioArchive, Member of the German Center for Lung Research (DZL) , Munich , Germany
| | - Gerald Burgstaller
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the CPC-M BioArchive, Member of the German Center for Lung Research (DZL) , Munich , Germany
| | - Jürgen Behr
- Asklepios Fachkliniken München-Gauting, Munich , Germany.,Medizinische Klinik und Poliklinik V, Klinikum der Ludwig-Maximilians-Universität, Munich , Germany
| | - Oliver Eickelberg
- Comprehensive Pneumology Center, University Hospital of the Ludwig-Maximilians-University Munich and Helmholtz Zentrum München, Member of the CPC-M BioArchive, Member of the German Center for Lung Research (DZL) , Munich , Germany.,Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado , Denver, Colorado
| |
Collapse
|
49
|
Woods K, Thigpen C, Wang JP, Park H, Hielscher A. Mechanically tuned 3 dimensional hydrogels support human mammary fibroblast growth and viability. BMC Cell Biol 2017; 18:35. [PMID: 29246104 PMCID: PMC5732527 DOI: 10.1186/s12860-017-0151-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 12/07/2017] [Indexed: 12/15/2022] Open
Abstract
Background Carcinoma associated fibroblasts (CAFs or myofibroblasts) are activated fibroblasts which participate in breast tumor growth, angiogenesis, invasion, metastasis and therapy resistance. As such, recent efforts have been directed toward understanding the factors responsible for activation of the phenotype. In this study, we have investigated how changes in the mechanical stiffness of a 3D hydrogel alter the behavior and myofibroblast-like properties of human mammary fibroblasts (HMFs). Results Here, we utilized microbial transglutaminase (mTG) to mechanically tune the stiffness of gelatin hydrogels and used rheology to show that increasing concentrations mTG resulted in hydrogels with greater elastic moduli (G’). Upon encapsulation of HMFs in 200 (compliant), 300 (moderate) and 1100 Pa (stiff) mTG hydrogels, it was found that the HMFs remained viable and proliferated over the 7 day culture period. Specifically, rates of proliferation were greatest for HMFs in moderate hydrogels. Regarding morphology, HMFs in compliant and moderate hydrogels exhibited a spindle-like morphology while HMFs in stiff hydrogels exhibited a rounded morphology with several large cellular protrusions. Quantification of cell morphology revealed that HMFs cultured in all mTG hydrogels overall assumed a more elongated phenotype over time in culture; however, few significant differences in morphology were observed between HMFs in each of the hydrogel conditions. To determine whether matrix stiffness upregulated expression of ECM and myofibroblast markers, western blot was performed on HMFs in compliant, moderate and stiff hydrogels. It was found that ECM and myofibroblast proteins varied in expression during both the culture period and according to matrix stiffness with no clear correlation between matrix stiffness and a myofibroblast phenotype. Finally, TGF-β levels were quantified in the conditioned media from HMFs in compliant, moderate and stiff hydrogels. TGF-β was significantly greater for HMFs encapsulated in stiff hydrogels. Conclusions Overall, these results show that while HMFs are viable and proliferate in mTG hydrogels, increasing matrix stiffness of mTG gelatin hydrogels doesn’t support a robust myofibroblast phenotype from HMFs. These results have important implications for further understanding how modulating 3D matrix stiffness affects fibroblast morphology and activation into a myofibroblast phenotype. Electronic supplementary material The online version of this article (10.1186/s12860-017-0151-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kathryn Woods
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Catlyn Thigpen
- Department of Biomedical Sciences, Georgia Philadelphia College of Osteopathic Medicine, Suwanee, GA, 30024, USA
| | - Jennifer Peyling Wang
- Anschutz Medical Campus Skaggs School of Pharmacy, University of Colorado, 12850 E. Montview Blvd, Aurora, CO, 80011, USA
| | - Hana Park
- Department of Biomedical Sciences, Georgia Philadelphia College of Osteopathic Medicine, Suwanee, GA, 30024, USA
| | - Abigail Hielscher
- Department of Biomedical Sciences, Georgia Philadelphia College of Osteopathic Medicine, Suwanee, GA, 30024, USA.
| |
Collapse
|
50
|
Bernau K, Torr EE, Evans MD, Aoki JK, Ngam CR, Sandbo N. Tensin 1 Is Essential for Myofibroblast Differentiation and Extracellular Matrix Formation. Am J Respir Cell Mol Biol 2017; 56:465-476. [PMID: 28005397 DOI: 10.1165/rcmb.2016-0104oc] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Myofibroblasts, the primary effector cells that mediate matrix remodeling during pulmonary fibrosis, rapidly assemble an extracellular fibronectin matrix. Tensin (TNS) 1 is a key component of specialized cellular adhesions (fibrillar adhesions) that bind to extracellular fibronectin fibrils. We hypothesized that TNS1 may play a role in modulating myofibroblast-mediated matrix formation. We found that TNS1 expression is increased in fibroblastic foci from lungs with idiopathic pulmonary fibrosis. Transforming growth factor (TGF)-β profoundly up-regulates TNS1 expression with kinetics that parallel the expression of the myofibroblast marker, smooth muscle α-actin. TGF-β-induced TNS1 expression is dependent on signaling through the TGF-β receptor 1 and is Rho coiled-coiled kinase/actin/megakaryoblastic leukemia-1/serum response factor dependent. Small interfering RNA-mediated knockdown of TNS1 disrupted TGF-β-induced myofibroblast differentiation, without affecting TGF-β/Smad signaling. In contrast, loss of TNS1 resulted in disruption of focal adhesion kinase phosphorylation, focal adhesion formation, and actin stress fiber development. Finally, TNS1 was essential for the formation of fibrillar adhesions and the assembly of nascent fibronectin and collagen matrix in myofibroblasts. In summary, our data show that TNS1 is a novel megakaryoblastic leukemia-1-dependent gene that is induced during pulmonary fibrosis. TNS1 plays an essential role in TGF-β-induced myofibroblast differentiation and myofibroblast-mediated formation of extracellular fibronectin and collagen matrix. Targeted disruption of TNS1 and associated signaling may provide an avenue to inhibit tissue fibrosis.
Collapse
Affiliation(s)
| | | | - Michael D Evans
- 2 Biostatistics and Medical Informatics, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin
| | | | | | | |
Collapse
|