2001
|
Varaprasad K, Jayaramudu T, Kanikireddy V, Toro C, Sadiku ER. Alginate-based composite materials for wound dressing application:A mini review. Carbohydr Polym 2020; 236:116025. [PMID: 32172843 DOI: 10.1016/j.carbpol.2020.116025] [Citation(s) in RCA: 380] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/03/2020] [Accepted: 02/16/2020] [Indexed: 12/12/2022]
Abstract
Alginate biopolymer has been used in the design and development of several wound dressing materials in order to improve the efficiency of wound healing. Mainly, alginate improves the hydrophilic nature of wound dressing materials in order to create the required moist wound environment, remove wound exudate and increase the speed of skin recovery of the wound. In addition, alginate can easily cross-link with other organic and inorganic materials and they can promote wound healing in clinical applications. This review article addresses the importance of alginates and the roles of derivative polymeric materials in wound dressing biomaterials. Additionally, studies on recent alginate-based wound dressing materials are discussed.
Collapse
Affiliation(s)
- Kokkarachedu Varaprasad
- Centro de Investigación de Polímeros Avanzados, CIPA, Avenida Collao 1202, Edificio de Laboratorios, Concepción, Chile.
| | - Tippabattini Jayaramudu
- Laboratory of Material Sciences, Instituto de Quimica de Recursos Naturales, Universidad de Talca, 747, Talca, Chile
| | - Vimala Kanikireddy
- Department of Chemistry, Osmania University, Hyderabad, 500 007, Telangana, India
| | - Claudio Toro
- Centro de Investigación de Polímeros Avanzados, CIPA, Avenida Collao 1202, Edificio de Laboratorios, Concepción, Chile
| | - Emmanuel Rotimi Sadiku
- Institute of NanoEngineering Research (INER), Department of Chemical, Metallurgical & Materials Engineering, (Polymer Division), Tshwane University of Technology, Pretoria West Campus, Staatsartillerie Rd, Pretoria, 0183, South Africa
| |
Collapse
|
2002
|
Kania G, Rudnik M, Distler O. Involvement of the myeloid cell compartment in fibrogenesis and systemic sclerosis. Nat Rev Rheumatol 2020; 15:288-302. [PMID: 30953037 DOI: 10.1038/s41584-019-0212-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Systemic sclerosis (SSc) is an autoimmune fibrotic disease of unknown aetiology that is characterized by vascular changes in the skin and visceral organs. Autologous haematopoietic stem cell transplantation can improve skin and organ fibrosis in patients with progressive disease and a high risk of organ failure, indicating that cells originating in the bone marrow are important contributors to the pathogenesis of SSc. Animal studies also indicate a pivotal function of myeloid cells in the development of fibrosis leading to changes in the tissue architecture and dysfunction in multiple organs such as the heart, lungs, liver and kidney. In this Review, we summarize current knowledge about the function of myeloid cells in fibrogenesis that occurs in patients with SSc. Targeted therapies currently in clinical studies for SSc might affect myeloid cell-related pathways. Therefore, myeloid cells might be used as cellular biomarkers of disease through the application of high-dimensional techniques such as mass cytometry and single-cell RNA sequencing.
Collapse
Affiliation(s)
- Gabriela Kania
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Michal Rudnik
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Oliver Distler
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
2003
|
Ma L, Li X, Guo X, Jiang Y, Li X, Guo H, Zhang B, Xu Y, Wang X, Li Q. Promotion of Endothelial Cell Adhesion and Antithrombogenicity of Polytetrafluoroethylene by Chemical Grafting of Chondroitin Sulfate. ACS APPLIED BIO MATERIALS 2020; 3:891-901. [PMID: 35019291 DOI: 10.1021/acsabm.9b00970] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Polytetrafluoroethylene (PTFE) is one of the polymers extensively applied in biomedicine. However, the application of PTFE as a small-diameter vascular graft results in thrombosis and intimal hyperplasia because of the immune response. Therefore, improving the biocompatibility and anticoagulant properties of PTFE is a key to solving this problem. In this study, a hydroxyl group-rich surface was obtained by oxidizing a benzoin-reduced PTFE membrane. Then, chondroitin sulfate (CS), an anticoagulant, was grafted on the surface of the hydroxylated PTFE membrane using 3-aminopropyltriethoxysilane. The successful modification of the membrane in each step was demonstrated by Fourier transform infrared spectroscopy and X-ray photoelectron spectroscopy. Hydroxylation and the grafting of CS greatly increased the hydrophilicity and roughness of membrane samples. Moreover, the hydroxylated PTFE membrane enhanced the adhesion ability of endothelial cells, and the grafting of CS also promoted the proliferation of endothelial cells and decreased platelet adhesion. The results indicate that the PTFE membranes grafted with CS are able to facilitate rapid endothelialization and inhibit thrombus formation, which makes the proposed method outstanding for artificial blood vessel applications.
Collapse
Affiliation(s)
- Lei Ma
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.,School of Materials Science & Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xuyan Li
- School of Materials Science & Engineering, Zhengzhou University, Zhengzhou 450001, China.,School of Mechanics & Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xin Guo
- School of Materials Science & Engineering, Zhengzhou University, Zhengzhou 450001, China.,School of Mechanics & Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Yongchao Jiang
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.,School of Materials Science & Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - XiaoMeng Li
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.,School of Mechanics & Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Haiyang Guo
- School of Materials Science & Engineering, Zhengzhou University, Zhengzhou 450001, China.,School of Mechanics & Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Bo Zhang
- School of Materials Science & Engineering, Zhengzhou University, Zhengzhou 450001, China.,School of Mechanics & Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Yiyang Xu
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.,School of Materials Science & Engineering, Zhengzhou University, Zhengzhou 450001, China.,Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin 53715, United States
| | - Xiaofeng Wang
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.,School of Mechanics & Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Qian Li
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.,School of Materials Science & Engineering, Zhengzhou University, Zhengzhou 450001, China.,School of Mechanics & Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
2004
|
Wen M, Zhi D, Wang L, Cui C, Huang Z, Zhao Y, Wang K, Kong D, Yuan X. Local Delivery of Dual MicroRNAs in Trilayered Electrospun Grafts for Vascular Regeneration. ACS APPLIED MATERIALS & INTERFACES 2020; 12:6863-6875. [PMID: 31958006 DOI: 10.1021/acsami.9b19452] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Globally growing problems related to cardiovascular diseases lead to a considerable need for synthetic vascular grafts. For small-caliber vascular prosthesis, it remains essential to fulfill rapid endothelialization, inhibit intimal hyperplasia, and prevent calcification for keeping patency. To modulate vascular regeneration, herein, we developed a bioactive trilayered tissue-engineered vascular graft encapsulating both microRNA-126 and microRNA-145 in the fibrous inner and middle layers, respectively. In vitro cell activities demonstrated that the trilayered electrospun membranes had significant biological advantages in enhanced growth and intracellular nitric oxide production of vascular endothelial cells, modulation of phenotypes of vascular smooth muscle cells (SMCs), and restraint of calcium deposition through fast-releasing microRNA-126 and slow-releasing microRNA-145. Histological and immunofluorescent analyses of in vivo implantation in a rat abdominal aorta interposition model suggested that the dual-microRNA-loading trilayered electrospun graft exerted a positive effect on accelerating endothelialization, improving contractile SMC regeneration, and promoting normal extracellular matrix formation. Meanwhile, the local bioactivity of microRNA-126 and microRNA-145 in the trilayered vascular graft could regulate inflammation and depress calcification possibly by facilitating transformation of macrophages into the anti-inflammatory M2 phenotype. These findings indicated that the trilayered electrospun graft by local delivery of dual microRNAs could be possibly used as a bioactive substitute for replacement of artificial small-caliber blood vessels.
Collapse
Affiliation(s)
- Meiling Wen
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Dengke Zhi
- Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences , Nankai University , Tianjin 300071 , China
| | - Lina Wang
- Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences , Nankai University , Tianjin 300071 , China
| | - Ce Cui
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Ziqi Huang
- Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences , Nankai University , Tianjin 300071 , China
| | - Yunhui Zhao
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| | - Kai Wang
- Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences , Nankai University , Tianjin 300071 , China
| | - Deling Kong
- Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Sciences , Nankai University , Tianjin 300071 , China
| | - Xiaoyan Yuan
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials , Tianjin University , Tianjin 300350 , China
| |
Collapse
|
2005
|
Brezovakova V, Jadhav S. Identification of Lyve-1 positive macrophages as resident cells in meninges of rats. J Comp Neurol 2020; 528:2021-2032. [PMID: 32003471 DOI: 10.1002/cne.24870] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 01/13/2023]
Abstract
Meningeal immunity along with its associated lymphatic vasculatures is widely discussed recently. Lymphatic vessels in meninges drain interstitial fluid into the deep-cervical lymph nodes. The vessels are composed of cells that express the cardinal marker for lymphatic endothelium-the lymphatic vessel hyaluronan receptor-1 (Lyve-1). However, studies also show the presence of nonendothelial Lyve-1 expressing cells in certain tissues. Therefore, we were curious if nonendothelial Lyve-1+ cells are also present in dura mater of meninges. We show that Lyve-1+ endothelial cells are distributed adjacent to the blood vessels in the brain dura mater of rats. We did not observe any lymphatic vessels in spinal dura mater. Interestingly, we also observed isolated population of nonlymphatic Lyve-1+ cells in both brain and spinal dura mater. Morphologically, the Lyve-1+ cells were extensively pleomorphic, sometimes elongated or round. Surprisingly, the thoracolumbal meningeal Lyve-1+ cells were predominantly round in morphology. Using endothelial specific marker VEGFR3 and macrophage markers CD68 and CD169, we observed that the isolated Lyve-1+ cells lacked endothelial cell signature, but were either CD68+ or CD169+ macrophages. Moreover, we observed that the Lyve-1+ cells colocalized with collagen fibers in the meninges, and some of Lyve-1+ cells had intracellular collagen. The study for the first time demonstrates the presence of Lyve-1 positive macrophages in the lymphatic and nonlymphatic regions in the meninges of rats.
Collapse
Affiliation(s)
- Veronika Brezovakova
- Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Institute of Neuroimmunology, Bratislava, Slovakia
| | - Santosh Jadhav
- Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Institute of Neuroimmunology, Bratislava, Slovakia
| |
Collapse
|
2006
|
Yang B, Chen Y, Li Z, Tang P, Tang Y, Zhang Y, Nie X, Fang C, Li X, Zhang H. Konjac glucomannan/polyvinyl alcohol nanofibers with enhanced skin healing properties by improving fibrinogen adsorption. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110718. [PMID: 32204030 DOI: 10.1016/j.msec.2020.110718] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/13/2020] [Accepted: 02/02/2020] [Indexed: 12/18/2022]
Abstract
Skin tissue engineering aims to develop the effective healing strategy to repair the wound by optimizing skin scaffold materials. During the skin wound healing process, fibrin plays an important role due to the specific blood coagulation effect. In this study, the outstanding fibrin capability of konjac glucomannan (KGM) is demonstrated by the molecular dynamics simulation and confirmed by the protein adsorption experiments. A series of konjac glucomannan/polyvinyl alcohol (KGM/PVA) composites with different ratio are fabricated and their role in enhancing the skin repair is tested by in vitro cell culture and in vivo study. The Eads (adsorption energy) between fibrin and KGM is about 30% larger than that between fibrin and PVA. The fibrinogen adsorption rates of PVA and KGM/PVA (5:5) composites can reach about 20% and 60%, respectively. The results show the blood adsorption capacity of KGM/PVA (5:5) composite can reach about 13 g/g. After 7 days of cell culture, the optical density values of 3T3 fibroblasts on KGM/PVA (5:5) composite could reach 0.8. The mechanical properties of the composites are also verified to meet the practical needs. Thus, we propose a potential wound dressing material strategy based on the materials design and the intrinsic properties of KGM.
Collapse
Affiliation(s)
- Bo Yang
- State Key Laboratory of Environmental Friendly Energy Materials, Engineering Research Center of Biomass Materials, Ministry of Education, School of Materials Science and Engineering, Southwest University of Science and Technology, Sichuan 621010, China
| | - Yushan Chen
- State Key Laboratory of Environmental Friendly Energy Materials, Engineering Research Center of Biomass Materials, Ministry of Education, School of Materials Science and Engineering, Southwest University of Science and Technology, Sichuan 621010, China
| | - Zhiqiang Li
- Department of Orthopedics, General Hospital of Western Theater Command, Chengdu 610038, China
| | - Pengfei Tang
- State Key Laboratory of Environmental Friendly Energy Materials, Engineering Research Center of Biomass Materials, Ministry of Education, School of Materials Science and Engineering, Southwest University of Science and Technology, Sichuan 621010, China
| | - Youhong Tang
- Institute for NanoScale Science and Technology and College of Science and Engineering, Flinders University, South Australia 5042, Australia
| | - Yaping Zhang
- State Key Laboratory of Environmental Friendly Energy Materials, Engineering Research Center of Biomass Materials, Ministry of Education, School of Materials Science and Engineering, Southwest University of Science and Technology, Sichuan 621010, China
| | - Xiaoqing Nie
- Fundamental Science on Nuclear Wastes and Environmental Safety Laboratory, Southwest University of Science and Technology, Mianyang 621010, China
| | - Cheng Fang
- Global Centre for Environmental Remediation, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Xiaodong Li
- Institute of Chemical Materials, China Academy of Engineering Physics (CAEP), Mianyang 621900, China.
| | - Hongping Zhang
- State Key Laboratory of Environmental Friendly Energy Materials, Engineering Research Center of Biomass Materials, Ministry of Education, School of Materials Science and Engineering, Southwest University of Science and Technology, Sichuan 621010, China.
| |
Collapse
|
2007
|
Wang L, Zhao Y, Yang F, Feng M, Zhao Y, Chen X, Mi J, Yao Y, Guan D, Xiao Z, Chen B, Dai J. Biomimetic collagen biomaterial induces in situ lung regeneration by forming functional alveolar. Biomaterials 2020; 236:119825. [PMID: 32044576 DOI: 10.1016/j.biomaterials.2020.119825] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/07/2020] [Accepted: 01/25/2020] [Indexed: 01/02/2023]
Abstract
In situ restoration of severely damaged lung remains difficult due to its limited regeneration capacity after injury. Artificial lung scaffolds are emerging as potential substitutes, but it is still a challenge to reconstruct lung regeneration microenvironment in scaffold after lung resection injury. Here, a 3D biomimetic porous collagen scaffold with similar structure characteristics as lung is fabricated, and a novel collagen binding hepatocyte growth factor (CBD-HGF) is tethered on the collagen scaffold for maintaining the biomimetic function of HGF to improve the lung regeneration microenvironment. The biomimetic scaffold was implanted into the operative region of a rat partial lung resection model. The results revealed that vascular endothelial cells and endogenous alveolar stem cells entered the scaffold at the early stage of regeneration. At the later stage, inflammation and fibrosis were attenuated, the microvascular and functional alveolar-like structures were formed, and the general morphology of the injured lung was restored. Taken together, the functional 3D biomimetic collagen scaffold facilitates recovery of the injured lung, alveolar regeneration, and angiogenesis after acute lung injury. Particularly, this is the first study of lung regeneration in vivo guided by biomimetic collagen scaffold materials, which supports the concept that tissue engineering is an effective strategy for alveolar regeneration.
Collapse
Affiliation(s)
- Linjie Wang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yannan Zhao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Feng Yang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Meng Feng
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yazhen Zhao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xi Chen
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Junwei Mi
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yuanjiang Yao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Dongwei Guan
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Zhifeng Xiao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Chen
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jianwu Dai
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
2008
|
Wang Y, Ma B, Yin A, Zhang B, Luo R, Pan J, Wang Y. Polycaprolactone vascular graft with epigallocatechin gallate embedded sandwiched layer-by-layer functionalization for enhanced antithrombogenicity and anti-inflammation. J Control Release 2020; 320:226-238. [PMID: 31982435 DOI: 10.1016/j.jconrel.2020.01.043] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 01/18/2020] [Accepted: 01/21/2020] [Indexed: 11/15/2022]
Abstract
Small-diameter artificial vascular grafts modified with layer-by-layer (LBL) coating show promise in reducing the failure caused by thrombosis and inflammation, but undesirable stability and bioactivity issues of the coating and payload usually limits their long-term efficacy. Herein, inspired by catechol/gallol surface chemistry, a sandwiched layer-by-layer coating constructed by polyethyleneimine (PEI) and heparin with the embedding of epigallocatechin gallate (EGCG)-dexamethasone combination was used to modify the electrospun polycaprolactone (PCL) vascular grafts. Polyphenol embedding endowed the coating with abundant intermolecular interactions between each coating components, mainly contributed by the π-π stacking, weak intermolecular cross-linking and enriched hydrogen bonding, which further enhanced the coating stability and also supported the sustained release of the payloads, like polyelectrolytes and drugs. Compared with the conventional LBL coating, the loading amounts of heparin and dexamethasone in the EGCG embedded LBL coatings doubled and the drug release could be significantly prolonged without serious initial burst. The in vitro and ex vivo assays indicated that the modified PCL vascular grafts would address impressive prolonged anti-platelet adhesion/activation and anti-fibrinogen denaturation ability. Meanwhile, the dexamethasone loading entrusted the sandwiched LBL coating with mild tissue response, in terms of inhibiting the macrophage activation. These results strongly demonstrated that the sandwiched LBL coating with EGCG embedding was an effective method to improve the patency rates of PCL small artificial vascular grafts, which could also be extended to other blood-contacting materials.
Collapse
Affiliation(s)
- Yanan Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Boxuan Ma
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Anlin Yin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Bo Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China.
| | - Junqiang Pan
- Department of Cardiovascular Medicine, Xi'an Central Hospital, Xi'an 710003, China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China.
| |
Collapse
|
2009
|
Jiang D, Rinkevich Y. Scars or Regeneration?-Dermal Fibroblasts as Drivers of Diverse Skin Wound Responses. Int J Mol Sci 2020; 21:E617. [PMID: 31963533 PMCID: PMC7014275 DOI: 10.3390/ijms21020617] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/13/2022] Open
Abstract
Scarring and regeneration are two physiologically opposite endpoints to skin injuries, with mammals, including humans, typically healing wounds with fibrotic scars. We aim to provide an updated review on fibroblast heterogeneity as determinants of the scarring-regeneration continuum. We discuss fibroblast-centric mechanisms that dictate scarring-regeneration continua with a focus on intercellular and cell-matrix adhesion. Improved understanding of fibroblast lineage-specific mechanisms and how they determine scar severity will ultimately allow for the development of antiscarring therapies and the promotion of tissue regeneration.
Collapse
Affiliation(s)
| | - Yuval Rinkevich
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Max-Lebsche-Platz 31, 81377 Munich, Germany;
| |
Collapse
|
2010
|
Tseng V, Ni K, Allawzi A, Prohaska C, Hernandez-Lagunas L, Elajaili H, Cali V, Midura R, Hascall V, Triggs-Raine B, Petrache I, Hart CM, Nozik-Grayck E. Extracellular Superoxide Dismutase Regulates Early Vascular Hyaluronan Remodeling in Hypoxic Pulmonary Hypertension. Sci Rep 2020; 10:280. [PMID: 31937874 PMCID: PMC6959284 DOI: 10.1038/s41598-019-57147-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/23/2019] [Indexed: 11/29/2022] Open
Abstract
Chronic hypoxia leads to pathologic remodeling of the pulmonary vasculature and pulmonary hypertension (PH). The antioxidant enzyme extracellular superoxide dismutase (SOD3) protects against hypoxia-induced PH. Hyaluronan (HA), a ubiquitous glycosaminoglycan of the lung extracellular matrix, is rapidly recycled at sites of vessel injury and repair. We investigated the hypothesis that SOD3 preserves HA homeostasis by inhibiting oxidative and enzymatic hyaluronidase-mediated HA breakdown. In SOD3-deficient mice, hypoxia increased lung hyaluronidase expression and activity, hyaluronan fragmentation, and effacement of HA from the vessel wall of small pulmonary arteries. Hyaluronan fragmentation corresponded to hypoxic induction of the cell surface hyaluronidase-2 (Hyal2), which was localized in the vascular media. Human pulmonary artery smooth muscle cells (HPASMCs) demonstrated hypoxic induction of Hyal2 and SOD-suppressible hyaluronidase activity, congruent to our observations in vivo. Fragmentation of homeostatic high molecular weight HA promoted HPASMC proliferation in vitro, whereas pharmacologic inhibition of hyaluronidase activity prevented hypoxia- and oxidant-induced proliferation. Hypoxia initiates SOD3-dependent alterations in the structure and regulation of hyaluronan in the pulmonary vascular extracellular matrix. These changes occurred soon after hypoxia exposure, prior to appearance of PH, and may contribute to the early pathogenesis of this disease.
Collapse
Affiliation(s)
- Victor Tseng
- Emory University Department of Medicine, Atlanta, GA, USA
| | - Kevin Ni
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ayed Allawzi
- Cardiovascular and Pulmonary Research, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Clare Prohaska
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Laura Hernandez-Lagunas
- Cardiovascular and Pulmonary Research, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hanan Elajaili
- Cardiovascular and Pulmonary Research, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Valbona Cali
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ronald Midura
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Vincent Hascall
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Barbara Triggs-Raine
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | - Irina Petrache
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - C Michael Hart
- Emory University Department of Medicine, Atlanta, GA, USA
| | - Eva Nozik-Grayck
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Cardiovascular and Pulmonary Research, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
2011
|
Feng Y, Wang Q, He M, Zhao W, Liu X, Zhao C. Nonadherent Zwitterionic Composite Nanofibrous Membrane with a Halloysite Nanocarrier for Sustained Wound Anti-Infection and Cutaneous Regeneration. ACS Biomater Sci Eng 2020; 6:621-633. [PMID: 33463235 DOI: 10.1021/acsbiomaterials.9b01547] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Wound dressing synechia and sustained postoperative bacterial infection would cause serious secondary damage to nascent cutaneous tissue and impede normal regeneration of injured wound. Endowing wound dressings with nonadherent capability and long-lasting antibacterial property could optimize the postoperative wound healing conditions and promote wound tissue neogenesis, which have important clinical application value and demand. In this study, novel nanocarrier-embedded zwitterionic composite nanofibrous membranes are fabricated using the co-electrospinning/photo-cross-linking method for the purpose of painless removal and eliminating long-lasting antibacterial infection during postoperative wound therapy. The prepared membranes possess good biocompatibility, excellent antibiofouling ability against both bacteria and plasma proteins, and platelet and L929 cell adhesion. Furthermore, in vitro and in vivo antibacterial evaluations exhibit that the composite nanofibrous membranes with a sustained drug release profile could effectively inhibit bacterial proliferation for at least 16 days. Additionally, in vivo wound regeneration assessment indicates that the obtained membranes could better enhance skin regeneration than the commercial 3M Tegaderm film, which highlights the application prospect of such novel zwitterionic composite nanofibrous membranes for sustained postoperative wound anti-infection and cutaneous regeneration.
Collapse
Affiliation(s)
- Yunbo Feng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Qian Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Min He
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Weifeng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Xiaoling Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Changsheng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| |
Collapse
|
2012
|
Zhao Y, Zhao X, Zhang R, Huang Y, Li Y, Shan M, Zhong X, Xing Y, Wang M, Zhang Y, Zhao Y. Cartilage Extracellular Matrix Scaffold With Kartogenin-Encapsulated PLGA Microspheres for Cartilage Regeneration. Front Bioeng Biotechnol 2020; 8:600103. [PMID: 33363129 PMCID: PMC7756004 DOI: 10.3389/fbioe.2020.600103] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022] Open
Abstract
Repair of articular cartilage defects is a challenging aspect of clinical treatment. Kartogenin (KGN), a small molecular compound, can induce the differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) into chondrocytes. Here, we constructed a scaffold based on chondrocyte extracellular matrix (CECM) and poly(lactic-co-glycolic acid) (PLGA) microspheres (MP), which can slowly release KGN, thus enhancing its efficiency. Cell adhesion, live/dead staining, and CCK-8 results indicated that the PLGA(KGN)/CECM scaffold exhibited good biocompatibility. Histological staining and quantitative analysis demonstrated the ability of the PLGA(KGN)/CECM composite scaffold to promote the differentiation of BMSCs. Macroscopic observations, histological tests, and specific marker analysis showed that the regenerated tissues possessed characteristics similar to those of normal hyaline cartilage in a rabbit model. Use of the PLGA(KGN)/CECM scaffold may mimic the regenerative microenvironment, thereby promoting chondrogenic differentiation of BMSCs in vitro and in vivo. Therefore, this innovative composite scaffold may represent a promising approach for acellular cartilage tissue engineering.
Collapse
Affiliation(s)
- Yanhong Zhao
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
- *Correspondence: Yanhong Zhao,
| | - Xige Zhao
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Rui Zhang
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Ying Huang
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Yunjie Li
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Minhui Shan
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Xintong Zhong
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Yi Xing
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | - Min Wang
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Medical University, Tianjin, China
| | | | - Yanmei Zhao
- Institute of Disaster Medicine, Tianjin University, Tianjin, China
- Yanmei Zhao,
| |
Collapse
|
2013
|
Liu J, Li B, Jing H, Wu Y, Kong D, Leng X, Wang Z. Swim Bladder as a Novel Biomaterial for Cardiovascular Materials with Anti-Calcification Properties. Adv Healthc Mater 2020; 9:e1901154. [PMID: 31815367 DOI: 10.1002/adhm.201901154] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/18/2019] [Indexed: 12/13/2022]
Abstract
Calcification is a major cause of cardiovascular materials failure and deterioration, which leads to the restriction of their wide application. To develop new materials with anti-calcification capability is an urgent clinical requirement. Herein, a natural material derived from swim bladders as one promising candidate is introduced, which is prepared by decellularization and glutaraldehyde (GA) crosslinking. Data show that the swim bladder is mainly composed of collagen I, glycosaminoglycan (GAG), and elastin, especially rich in elastin, in accordance with higher elastic modulus in comparison to bovine pericardium. Moreover, the calcification of this material is proved dramatically lower than that of bovine pericardium by in vitro calcification assessments and in vivo assay using a rat subcutaneous implantation model. Meanwhile, good cytocompatibility, hemocompatibility, and enzymatic stability are demonstrated by in vitro assays. Further, a small diameter vascular graft using this material is successfully developed by rolling method and in situ implantation assay using a rat abdominal artery replacement model shows great performances in the aspect of higher patency and lower calcification. Taken together, these superior properties of swim bladder-derived material in anti-calcification, proper mechanical strength and stability, and excellent hemocompatibility and cytocompatibility endow it a great candidate as cardiovascular biomaterials.
Collapse
Affiliation(s)
- Jing Liu
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Binhan Li
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Huimin Jing
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Yongjian Wu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Deling Kong
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
- Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Xigang Leng
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Zhihong Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| |
Collapse
|
2014
|
Gao H, Zhong Z, Xia H, Hu Q, Ye Q, Wang Y, Chen L, Du Y, Shi X, Zhang L. Construction of cellulose nanofibers/quaternized chitin/organic rectorite composites and their application as wound dressing materials. Biomater Sci 2019; 7:2571-2581. [PMID: 30977470 DOI: 10.1039/c9bm00288j] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Traumatic injury is a major cause of mortality, and poor wound healing affects millions of people. Thus, the development of effective wound dressings is essential for speeding up wound healing and decreasing mortality. In this study, a suspension of carboxylated brown algae cellulose nanofibers (BACNFs) with a high aspect ratio was freeze dried to prepare a sponge. The sponge showed high porosity and water absorption capacity; thus, it can absorb wound exudates when used as a wound dressing. In addition, quaternized β-chitin (QC) with antibacterial properties was intercalated into the interlayer space of the organic rectorite (OREC) via electrostatic interactions to obtain composite suspensions (QCRs) with improved antimicrobial activity compared to that of QC alone. Subsequently, the BACNF sponge was soaked in the QCR suspension to absorb QCRs via electrostatic interactions and hydrogen bonding from which cellulose nanofiber/quaternized chitin/organic rectorite composite (BACNF/QCR) sponges were constructed via freeze-drying. The in vivo animal tests demonstrated that the BACNF/QCR sponges rapidly induced hemostasis in a rat tail amputation test, making them superior to the traditional hemostatic materials. Furthermore, BACNFs/QCRs could substantially promote collagen synthesis and neovascularization, thereby accelerating wound healing 3 days earlier than gauze. This multi-functional biomedical material, fabricated using natural substances, shows great potential to be used for wound healing.
Collapse
Affiliation(s)
- Huimin Gao
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
2015
|
Sasaki N, Toyoda M. Vascular Diseases and Gangliosides. Int J Mol Sci 2019; 20:ijms20246362. [PMID: 31861196 PMCID: PMC6941100 DOI: 10.3390/ijms20246362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/12/2019] [Accepted: 12/16/2019] [Indexed: 02/07/2023] Open
Abstract
Vascular diseases, such as myocardial infarction and cerebral infarction, are most commonly caused by atherosclerosis, one of the leading causes of death worldwide. Risk factors for atherosclerosis include lifestyle and aging. It has been reported that lifespan could be extended in mice by targeting senescent cells, which led to the suppression of aging-related diseases, such as vascular diseases. However, the molecular mechanisms underlying the contribution of aging to vascular diseases are still not well understood. Several types of cells, such as vascular (endothelial cell), vascular-associated (smooth muscle cell and fibroblast) and inflammatory cells, are involved in plaque formation, plaque rupture and thrombus formation, which result in atherosclerosis. Gangliosides, a group of glycosphingolipids, are expressed on the surface of vascular, vascular-associated and inflammatory cells, where they play functional roles. Clarifying the role of gangliosides in atherosclerosis and their relationship with aging is fundamental to develop novel prevention and treatment methods for vascular diseases based on targeting gangliosides. In this review, we highlight the involvement and possible contribution of gangliosides to vascular diseases and further discuss their relationship with aging.
Collapse
Affiliation(s)
- Norihiko Sasaki
- Correspondence: (N.S.); (M.T.); Tel.: +81-3-3964-3241 (N.S.); +81-3-3964-4421 (M.T.)
| | - Masashi Toyoda
- Correspondence: (N.S.); (M.T.); Tel.: +81-3-3964-3241 (N.S.); +81-3-3964-4421 (M.T.)
| |
Collapse
|
2016
|
Garoffolo G, Pesce M. Mechanotransduction in the Cardiovascular System: From Developmental Origins to Homeostasis and Pathology. Cells 2019; 8:cells8121607. [PMID: 31835742 PMCID: PMC6953076 DOI: 10.3390/cells8121607] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/04/2019] [Accepted: 12/10/2019] [Indexed: 12/16/2022] Open
Abstract
With the term ‘mechanotransduction’, it is intended the ability of cells to sense and respond to mechanical forces by activating intracellular signal transduction pathways and the relative phenotypic adaptation. While a known role of mechanical stimuli has been acknowledged for developmental biology processes and morphogenesis in various organs, the response of cells to mechanical cues is now also emerging as a major pathophysiology determinant. Cells of the cardiovascular system are typically exposed to a variety of mechanical stimuli ranging from compression to strain and flow (shear) stress. In addition, these cells can also translate subtle changes in biophysical characteristics of the surrounding matrix, such as the stiffness, into intracellular activation cascades with consequent evolution toward pro-inflammatory/pro-fibrotic phenotypes. Since cellular mechanotransduction has a potential readout on long-lasting modifications of the chromatin, exposure of the cells to mechanically altered environments may have similar persisting consequences to those of metabolic dysfunctions or chronic inflammation. In the present review, we highlight the roles of mechanical forces on the control of cardiovascular formation during embryogenesis, and in the development and pathogenesis of the cardiovascular system.
Collapse
Affiliation(s)
- Gloria Garoffolo
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Via Parea, 4, I-20138 Milan, Italy;
- PhD Program in Translational and Molecular Medicine DIMET, Università di Milano - Bicocca, 20126 Milan, Italy
- Correspondence:
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Via Parea, 4, I-20138 Milan, Italy;
| |
Collapse
|
2017
|
Mesenchymal Stem Cell-Derived Extracellular Vesicles for Corneal Wound Repair. Stem Cells Int 2019; 2019:5738510. [PMID: 31885617 PMCID: PMC6925772 DOI: 10.1155/2019/5738510] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/14/2019] [Accepted: 11/26/2019] [Indexed: 12/27/2022] Open
Abstract
With the immunoregulation potential, mesenchymal stem cells (MSCs) have been used for tissue regeneration by relieving inflammation in the injured tissues. When this repair process is interfered by immune disorders or pathological angiogenesis, the delays in corneal epithelial wound healing can lead to a persistent epithelial defect. Stem cell-derived extracellular vesicles (EVs), which carry abundant bioactive molecules from stem cells, have provided an alternative to regeneration therapy. In this study, we aimed to investigate if EVs from human placenta-derived MSCs (hP-MSCs) could ameliorate alkali injury of the cornea in the mouse model. 33.33 μg/μL EVs in 10 μL PBS were applied to the cornea. Repeat application three times, and 100 μg EVs (in 30 μL PBS) in total were administrated per day for two weeks. Our results revealed that EVs from hP-MSCs had preferable functions including enhancing proliferation and anti-inflammation and suppressing apoptosis of corneal epithelial cells. Furthermore, hP-MSC-derived EVs ameliorated mouse corneal wound healing by inhibiting angiogenesis and inflammation. Taken together, our current data suggested that hP-MSC-derived EVs have the beneficial effects of corneal wound healing, which provide alternative cell-free therapy with great practical value.
Collapse
|
2018
|
Li Y, Liu Y, Han X, Jin H, Ma S. Arsenic Species in Cordyceps sinensis and Its Potential Health Risks. Front Pharmacol 2019; 10:1471. [PMID: 31866869 PMCID: PMC6910106 DOI: 10.3389/fphar.2019.01471] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/13/2019] [Indexed: 01/20/2023] Open
Abstract
High arsenic residues make Cordyceps sinensis a concern in China. Arsenic toxicity is related to its species. Many studies have evaluated the toxicity of total arsenic, but few have studied its species. In this study, the species of arsenic in C. sinensis and its potential health risk were investigated. SEC-HPLC-ICP-MS was used to analysis of arsenic in C. sinensis and unknown arsenic (uAs) was discovered. Additionally, arsenic in C. sinensis was mainly found in alkali-soluble proteins. The trend of arsenic transformation indicated that unknown arsenic in C. sinensis may be converted into free inorganic arsenic, which enhanced toxicity. The result of risk assessment indicated that there were potential health risks of uAs. Hereon, we proposed recommendations for the use of C. sinensis and regulatory recommendations for arsenic standards. This study contributed to the toxicity reveal, safety evaluation, and risk assessment of arsenic in C. sinensis.
Collapse
Affiliation(s)
- Yaolei Li
- National Institutes for Food and Drug Control, Beijing, China
| | - Yue Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao Han
- Department of Pharmacy, Beihua University, Jilin, China
| | - Hongyu Jin
- National Institutes for Food and Drug Control, Beijing, China
| | - Shuangcheng Ma
- National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
2019
|
Richards CD, Botelho F. Oncostatin M in the Regulation of Connective Tissue Cells and Macrophages in Pulmonary Disease. Biomedicines 2019; 7:E95. [PMID: 31817403 PMCID: PMC6966661 DOI: 10.3390/biomedicines7040095] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/23/2019] [Accepted: 11/26/2019] [Indexed: 12/16/2022] Open
Abstract
Oncostatin M (OSM), as one of the gp130/IL-6 family of cytokines, interacts with receptor complexes that include the gp130 signaling molecule and OSM receptor β OSMRβ chain subunits. OSMRβ chains are expressed relatively highly across a broad array of connective tissue (CT) cells of the lung, such as fibroblasts, smooth muscle cells, and epithelial cells, thus enabling robust responses to OSM, compared to other gp130 cytokines, in the regulation of extracellular matrix (ECM) remodeling and inflammation. OSMRβ chain expression in lung monocyte/macrophage populations is low, whereas other receptor subunits, such as that for IL-6, are present, enabling responses to IL-6. OSM is produced by macrophages and neutrophils, but not CT cells, indicating a dichotomy of OSM roles in macrophage verses CT cells in lung inflammatory disease. ECM remodeling and inflammation are components of a number of chronic lung diseases that show elevated levels of OSM. OSM-induced products of CT cells, such as MCP-1, IL-6, and PGE2 can modulate macrophage function, including the expression of OSM itself, indicating feedback loops that characterize Macrophage and CT cell interaction.
Collapse
Affiliation(s)
- Carl D. Richards
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 3Z5, Canada;
| | | |
Collapse
|
2020
|
Lu L, Xie R, Wei R, Cai C, Bi D, Yin D, Liu H, Zheng J, Zhang Y, Song F, Gao Y, Tan L, Wei Q, Qin H. Integrin α5 subunit is required for the tumor supportive role of fibroblasts in colorectal adenocarcinoma and serves as a potential stroma prognostic marker. Mol Oncol 2019; 13:2697-2714. [PMID: 31600854 PMCID: PMC6887586 DOI: 10.1002/1878-0261.12583] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/24/2019] [Accepted: 10/08/2019] [Indexed: 12/18/2022] Open
Abstract
The tumorigenesis of colorectal cancer (CRC) is a complicated process, involving interactions between cancer cells and the microenvironment. The role of α5 integrin subunit in CRC remains controversial, and previous studies mainly focused on cancer cells. Herein, we report an important role of α5 in stroma fibroblasts in the tumorigenesis of CRC. The expression of α5 was found to be located in colorectal tumor stroma rather than in epithelia cancer cells. Immunofluorescence colocalization and gene correlation analysis confirmed that α5 was mainly expressed in cancer-associated fibroblasts (CAFs). Moreover, experimental evidence showed that α5 expression was required for the tumor-promoting effect of fibroblast cells. In an in vivo xenograft nude mice model, α5 depletion in fibroblasts dramatically suppressed fibroblast-induced tumor growth. In an in vitro cell coculture assay, α5 depletion or knockdown reduced the ability of fibroblasts to promote cancer cell migration and invasion compared with wild-type fibroblasts; moreover, we observed that the expression and assembly of fibronectin were downregulated after α5 depletion or knockdown in fibroblasts. Analysis of the RNA-Seq data of the Cancer Genome Atlas cohort revealed that high expression of ITGA5 (α5 integrin subunit) was correlated with poor overall survival in colorectal adenocarcinoma, which was further confirmed by immunohistochemistry in an independent cohort of 355 patients. Thus, our study identifies α5 integrin subunit as a novel stroma molecular marker for colorectal adenocarcinoma, offers a fresh insight into colorectal adenocarcinoma progression, and shows that α5 expression in stroma fibroblasts underlies its ability to promote the tumorigenesis of colorectal adenocarcinoma.
Collapse
Affiliation(s)
- Ling Lu
- Department of PathologyShanghai Tenth People’s Hospital Affiliated to Tongji UniversityChina
| | - Ruting Xie
- Department of PathologyShanghai Tenth People’s Hospital Affiliated to Tongji UniversityChina
| | - Rong Wei
- Department of PathologyShanghai Tenth People’s Hospital Affiliated to Tongji UniversityChina
| | - Chunmiao Cai
- Department of PathologyShanghai Tenth People’s Hospital Affiliated to Tongji UniversityChina
| | - Dexi Bi
- Department of PathologyShanghai Tenth People’s Hospital Affiliated to Tongji UniversityChina
| | - Dingzi Yin
- Department of Gastrointestinal SurgeryShanghai Tenth People’s Hospital Affiliated to Tongji UniversityChina
| | - Hu Liu
- Department of PathologyShanghai Tenth People’s Hospital Affiliated to Tongji UniversityChina
| | - Jiayi Zheng
- Department of PathologyShanghai Tenth People’s Hospital Affiliated to Tongji UniversityChina
| | - Youhua Zhang
- Department of PathologyShanghai Tenth People’s Hospital Affiliated to Tongji UniversityChina
| | - Feifei Song
- Department of PathologyShanghai Tenth People’s Hospital Affiliated to Tongji UniversityChina
| | - Yaohui Gao
- Department of PathologyShanghai Tenth People’s Hospital Affiliated to Tongji UniversityChina
| | - Linhua Tan
- Department of PathologyShanghai Tenth People’s Hospital Affiliated to Tongji UniversityChina
| | - Qing Wei
- Department of PathologyShanghai Tenth People’s Hospital Affiliated to Tongji UniversityChina
| | - Huanlong Qin
- Division of GastroenterologyDepartment of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| |
Collapse
|
2021
|
Kuse N, Kamio K, Azuma A, Matsuda K, Inomata M, Usuki J, Morinaga A, Tanaka T, Kashiwada T, Atsumi K, Hayashi H, Saito Y, Seike M, Gemma A. Exosome-Derived microRNA-22 Ameliorates Pulmonary Fibrosis by Regulating Fibroblast-to-Myofibroblast Differentiation in Vitro and in Vivo. J NIPPON MED SCH 2019; 87:118-128. [PMID: 31776321 DOI: 10.1272/jnms.jnms.2020_87-302] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Although aberrant proliferation and activation of lung fibroblasts are implicated in the initiation and progression of idiopathic pulmonary fibrosis (IPF), the underlying mechanisms are not well characterized. Numerous microRNAs (miRNAs) have been implicated in this process; however, miRNAs derived from exosomes and the relevance of such miRNAs to fibroblast-to-myofibroblast differentiation are not well understood. In this study, we attempted to identify exosome-derived miRNAs relevant to fibrosis development. METHODS Using miRNA array analysis, we profiled exosome-derived miRNA expression in sera of C57BL/6 mice exhibiting bleomycin-induced pulmonary fibrosis. After validating a selected miRNA by quantitative reverse-transcription polymerase chain reaction, its effect on fibroblast-to-myofibroblast differentiation was investigated in human lung fibroblasts. Furthermore, we determined the role of the selected miRNA in an in vivo model of pulmonary fibrosis. RESULTS MiRNA array analysis revealed that miR-22 expression was increased by up to 2 fold on day 7 after bleomycin treatment compared with that in vehicle-treated mice. In vitro, miR-22 transfection suppressed TGF-β1-induced α-SMA expression. This was mediated via inhibition of the ERK1/2 pathway. Baseline α-SMA expression was increased upon miR-22 inhibitor transfection. Furthermore, miR-22 negatively regulated connective tissue growth factor expression in the presence of TGF-β1. In vivo, administration of a miR-22 mimic on day 10 after bleomycin challenge ameliorated pulmonary fibrosis lesions accompanied by decreased α-SMA expression in the model mice. CONCLUSIONS Exosomal miR-22 modulates fibroblast-to-myofibroblast differentiation. The present findings warrant further study, which could shed light on miR-22 as a novel therapeutic target in IPF.
Collapse
Affiliation(s)
- Naoyuki Kuse
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School
| | - Koichiro Kamio
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School
| | - Arata Azuma
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School
| | - Kuniko Matsuda
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School
| | - Minoru Inomata
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School
| | - Jiro Usuki
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School
| | - Akemi Morinaga
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School
| | - Toru Tanaka
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School
| | - Takeru Kashiwada
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School
| | - Kenichiro Atsumi
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School
| | - Hiroki Hayashi
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School
| | - Yoshinobu Saito
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School
| | - Masahiro Seike
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School
| | - Akihiko Gemma
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School
| |
Collapse
|
2022
|
Azbazdar Y, Ozalp O, Sezgin E, Veerapathiran S, Duncan AL, Sansom MSP, Eggeling C, Wohland T, Karaca E, Ozhan G. More Favorable Palmitic Acid Over Palmitoleic Acid Modification of Wnt3 Ensures Its Localization and Activity in Plasma Membrane Domains. Front Cell Dev Biol 2019; 7:281. [PMID: 31803740 PMCID: PMC6873803 DOI: 10.3389/fcell.2019.00281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/31/2019] [Indexed: 12/17/2022] Open
Abstract
While the lateral organization of plasma membrane components has been shown to control binding of Wnt ligands to their receptors preferentially in the ordered membrane domains, the role of posttranslational lipid modification of Wnt on this selective binding is unknown. Here, we identify that the canonical Wnt is presumably acylated by palmitic acid, a saturated 16-carbon fatty acid, at a conserved serine residue. Acylation of Wnt3 is dispensable for its secretion and binding to Fz8 while it is essential for Wnt3's proper binding and domain-like diffusion in the ordered membrane domains. We further unravel that non-palmitoylated Wnt3 is unable to activate Wnt/β-catenin signaling either in zebrafish embryos or in mammalian cells. Based on these results, we propose that the lipidation of canonical Wnt, presumably by a saturated fatty acid, determines its competence in interacting with the receptors in the appropriate domains of the plasma membrane, ultimately keeping the signaling activity under control.
Collapse
Affiliation(s)
- Yagmur Azbazdar
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, Izmir, Turkey
| | - Ozgun Ozalp
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, Izmir, Turkey
| | - Erdinc Sezgin
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Sapthaswaran Veerapathiran
- Department of Biological Sciences and Center for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Anna L. Duncan
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Mark S. P. Sansom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Christian Eggeling
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Department of Super-Resolution Microscopy, Institute for Applied Optics and Biophysics, Friedrich-Schiller-University Jena, Jena, Germany
- Department of Biophysical Imaging, Leibniz Institute of Photonic Technology e.V., Jena, Germany
| | - Thorsten Wohland
- Department of Biological Sciences and Center for BioImaging Sciences, National University of Singapore, Singapore, Singapore
- Department of Chemistry, National University of Singapore, Singapore, Singapore
| | - Ezgi Karaca
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, Izmir, Turkey
| | - Gunes Ozhan
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, Izmir, Turkey
| |
Collapse
|
2023
|
Zhu T, Jiang J, Zhao J, Chen S, Yan X. Regulating Preparation Of Functional Alginate-Chitosan Three-Dimensional Scaffold For Skin Tissue Engineering. Int J Nanomedicine 2019; 14:8891-8903. [PMID: 32009786 PMCID: PMC6859126 DOI: 10.2147/ijn.s210329] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 10/21/2019] [Indexed: 12/14/2022] Open
Abstract
AIM In this study, we attempted to regulate the preparation of Alg-CS-Flu three-dimensional scaffolds via a facile freeze-drying method combined with amidation. MATERIALS AND METHODS Three-dimensional porous flurbiprofen-grafted alginate (Alg)-chitosan (CS) scaffolds were successfully prepared by a facile freeze-drying method combined with amidation for skin tissue engineering applications. Alg-CS composite was first used to load flurbiprofen (Flu), which is a kind of anti-inflammatory non-steroidal molecule. The Flu-loaded Alg/CS composite solution, through freeze-drying and 1-ethyl-3(3-(dimethylamino)propyl) carbodiimide/N-hydroxysuccinimide crosslinking to form an Alg-CS-Flu scaffold, exhibited a uniform and porous morphology that was characterized using scanning electron microscopy. The Alg-CS-Flu as-prepared scaffold was also characterized using Fourier-transform infrared spectroscopy, water contact angle, thermal properties, and stress-strain testing. RESULTS The results reveal that Flu was successfully grafted onto the surfaces of the Alg-CS-Flu scaffold, which showed good hydrophilicity and appropriate mechanical properties. Furthermore, cell viability, cell morphology from cells cultured in vitro, and hematoxylin-eosin staining after the graft was subcutaneously embedded in mice for 7 d demonstrated that the Alg-CS-Flu scaffold had no unfavorable effects on the adhesion and proliferation of fibroblasts, as well as a good anti-inflammatory property. CONCLUSION The developed Alg-CS-Flu scaffold is proposed as a promising material or skin tissue engineering application.
Collapse
Affiliation(s)
- Tonghe Zhu
- Department of Sports Medicine, Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai200233, People’s Republic of China
| | - Jia Jiang
- Department of Sports Medicine, Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai200233, People’s Republic of China
| | - Jinzhong Zhao
- Department of Sports Medicine, Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai200233, People’s Republic of China
| | - Sihao Chen
- Scientific Research Department of Shanghai University of Engineering Science, Shanghai201620, People’s Republic of China
- Multidisciplinary Center for Advanced Materials of Shanghai University of Engineering Science, Shanghai University of Engineering Science, Shanghai201620, People’s Republic of China
| | - Xiaoyu Yan
- Department of Sports Medicine, Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai200233, People’s Republic of China
| |
Collapse
|
2024
|
Wu Y, Zhao Y, He X, He Z, Wang T, Wan L, Chen L, Yan N. Hydroxypropyl‑β‑cyclodextrin attenuates the epithelial‑to‑mesenchymal transition via endoplasmic reticulum stress in MDA‑MB‑231 breast cancer cells. Mol Med Rep 2019; 21:249-257. [PMID: 31746388 PMCID: PMC6896369 DOI: 10.3892/mmr.2019.10802] [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: 04/26/2019] [Accepted: 10/02/2019] [Indexed: 01/04/2023] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) has been reported to serve vital roles in regulating the progress of cancer metastasis. In addition, lipid rafts enriched in sphingolipids and cholesterol serve important roles in physiological and biochemical processes as a signaling platform. The present study explored the effects of hydroxypropyl-β-cyclodextrin (HP-β-CD), a cholesterol-depleting agent of lipid rafts, on the transforming growth factor (TGF)-β/Smad signaling pathway and endoplasmic reticulum (ER) stress in mediating EMT in MDA-MB-231 breast cancer cells. HP-β-CD treatment inhibited TGF-β1-induced EMT, based on increased expression of E-cadherin and decreased expression of vimentin. HP-β-CD reduced the expression of the TGF receptor TβRI and blocked the phosphorylation of Smad2. In addition, HP-β-CD increased the expression of ER stress-related proteins (binding immunoglobulin protein and activating transcription factor 6), but TGF-β1 could reverse these changes. Sodium 4-phenylbutyrate, an inhibitor of ER stress, suppressed these effects of HP-β-CD on EMT and TGF-β/Smad signaling pathway inhibition in breast cancer cells. Thus, HP-β-CD can block the TGF-β/Smad signaling pathway via diminishing the expression of TβRI which helps to activate ER stress and attenuate EMT in MDA-MB-231 cells, highlighting a potential target of lipid rafts for breast cancer treatment.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yiyang Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xuanhong He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhiqiang He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tian Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Linxi Wan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lai Chen
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, P.R. China
| | - Nianlong Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
2025
|
Yuan H, Chen C, Liu Y, Lu T, Wu Z. Strategies in cell‐free tissue‐engineered vascular grafts. J Biomed Mater Res A 2019; 108:426-445. [PMID: 31657523 DOI: 10.1002/jbm.a.36825] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Haoyong Yuan
- Department of Cardiovascular surgery The Second Xiangya Hospital of Central South University Changsha Hunan China
| | - Chunyang Chen
- Department of Cardiovascular surgery The Second Xiangya Hospital of Central South University Changsha Hunan China
| | - Yuhong Liu
- Department of Cardiovascular surgery The Second Xiangya Hospital of Central South University Changsha Hunan China
| | - Ting Lu
- Department of Cardiovascular surgery The Second Xiangya Hospital of Central South University Changsha Hunan China
| | - Zhongshi Wu
- Department of Cardiovascular surgery The Second Xiangya Hospital of Central South University Changsha Hunan China
| |
Collapse
|
2026
|
Penke LR, Peters-Golden M. Molecular determinants of mesenchymal cell activation in fibroproliferative diseases. Cell Mol Life Sci 2019; 76:4179-4201. [PMID: 31563998 PMCID: PMC6858579 DOI: 10.1007/s00018-019-03212-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/01/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023]
Abstract
Uncontrolled scarring, or fibrosis, can interfere with the normal function of virtually all tissues of the body, ultimately leading to organ failure and death. Fibrotic diseases represent a major cause of death in industrialized countries. Unfortunately, no curative treatments for these conditions are yet available, highlighting the critical need for a better fundamental understanding of molecular mechanisms that may be therapeutically tractable. The ultimate indispensable effector cells responsible for deposition of extracellular matrix proteins that comprise scars are mesenchymal cells, namely fibroblasts and myofibroblasts. In this review, we focus on the biology of these cells and the molecular mechanisms that regulate their pertinent functions. We discuss key pro-fibrotic mediators, signaling pathways, and transcription factors that dictate their activation and persistence. Because of their possible clinical and therapeutic relevance, we also consider potential brakes on mesenchymal cell activation and cellular processes that may facilitate myofibroblast clearance from fibrotic tissue-topics that have in general been understudied.
Collapse
Affiliation(s)
- Loka R Penke
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109-5642, USA
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109-5642, USA.
| |
Collapse
|
2027
|
Liang Y, Zhao X, Hu T, Han Y, Guo B. Mussel-inspired, antibacterial, conductive, antioxidant, injectable composite hydrogel wound dressing to promote the regeneration of infected skin. J Colloid Interface Sci 2019; 556:514-528. [DOI: 10.1016/j.jcis.2019.08.083] [Citation(s) in RCA: 374] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 01/11/2023]
|
2028
|
Shih SR, Liao SL, Shih CW, Wei YH, Lu TX, Chou CH, Yen EY, Chang YC, Lin CC, Chi YC, Yang WS, Tsai FC. Fibroblast Growth Factor Receptor Inhibitors Reduce Adipogenesis of Orbital Fibroblasts and Enhance Myofibroblastic Differentiation in Graves' Orbitopathy. Ocul Immunol Inflamm 2019; 29:193-202. [PMID: 31657648 DOI: 10.1080/09273948.2019.1672196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Purpose: Orbital fibroblasts are involved in pathogenesis of Graves' orbitopathy (GO). Fibroblast growth factor (FGF) affects fibroblasts of GO. This study aims to investigate the roles of FGF and FGF receptor (FGFR) in GO.Methods: Serum FGF proteins and orbital fibroblast FGFR proteins and mRNAs were measured in GO patients and controls. Orbital fibroblasts of GO were cultured and accessed for changes in proliferation (by nuclei number and MTT), myofibroblastic differentiation (by α-SMA), and adipogenesis (by oil droplets using Oil Red O stain) under FGF1 with or without FGFR inhibitors (FGFRi).Results: Serum FGF1 and FGF2 were increased in GO patients. FGFR1 was the most abundantly expressed FGFR in GO orbital fibroblasts. FGF1 increased GO fibroblast proliferation/adipogenesis and suppressed myofibroblastic differentiation, while FGFRi reversed these effects.Conclusion: FGF signaling may be involved in GO pathogenesis. Manipulation of FGF-FGFR pathway for GO treatment is worthy of further investigation.Registration number on Clinicaltrials.gov: NCT03324022.
Collapse
Affiliation(s)
- Shyang-Rong Shih
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Center of Anti-Aging and Health Consultation, National Taiwan University Hospital, Taipei, Taiwan
| | - Shu-Lang Liao
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Ophthalmology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chih-Wei Shih
- Department of Ophthalmology, Zhongxing Branch, Taipei City Hospital, Taipei, Taiwan
| | - Yi-Hsuan Wei
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ting-Xuan Lu
- Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chien-Hsiang Chou
- Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Er-Yen Yen
- Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yi-Cheng Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan.,Institute of Biomedical Science, Academia Sinica, Taipei, Taiwan
| | - Chia-Chi Lin
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Chiao Chi
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wei-Shiung Yang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Center for Obesity, Lifestyle, and Metabolic Surgery, National Taiwan University Hospital, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Feng-Chiao Tsai
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
2029
|
Ghazvini HJ, Armaghan M, Janiak C, Balalaie S, Müller TJJ. Coupling‐Isomerization‐Cycloisomerization Reaction (CICIR) – An Unexpected and Efficient Domino Approach to Luminescent 2‐(Hydroxymethylene)indenones. European J Org Chem 2019. [DOI: 10.1002/ejoc.201901413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Helya Janatian Ghazvini
- Institut für Organische Chemie und Makromolekular Chemie Heinrich‐Heine‐Universität Düsseldorf Universitätsstraße 1 40225 Düsseldorf Germany
- Peptide Chemistry Research Center K. N. Toosi University of Technology P. O. Box 15875‐4416 Tehran Iran
| | - Mahsa Armaghan
- Institut für Anorganische Chemie und Strukturchemie Heinrich‐Heine‐Universität Düsseldorf Universitätsstraße 1 40225 Düsseldorf Germany
| | - Christoph Janiak
- Institut für Anorganische Chemie und Strukturchemie Heinrich‐Heine‐Universität Düsseldorf Universitätsstraße 1 40225 Düsseldorf Germany
| | - Saeed Balalaie
- Peptide Chemistry Research Center K. N. Toosi University of Technology P. O. Box 15875‐4416 Tehran Iran
| | - Thomas J. J. Müller
- Institut für Organische Chemie und Makromolekular Chemie Heinrich‐Heine‐Universität Düsseldorf Universitätsstraße 1 40225 Düsseldorf Germany
| |
Collapse
|
2030
|
Lin CY, Kolliopoulos C, Huang CH, Tenhunen J, Heldin CH, Chen YH, Heldin P. High levels of serum hyaluronan is an early predictor of dengue warning signs and perturbs vascular integrity. EBioMedicine 2019; 48:425-441. [PMID: 31526718 PMCID: PMC6838418 DOI: 10.1016/j.ebiom.2019.09.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/30/2019] [Accepted: 09/06/2019] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND A main pathological feature of severe dengue virus infection is endothelial hyper-permeability. The dengue virus nonstructural protein 1 (NS1) has been implicated in the vascular leakage that characterizes severe dengue virus infection, however, the molecular mechanisms involved are not known. METHODS A cohort of 250 dengue patients has been followed from the onset of symptoms to the recovery phase. Serum hyaluronan levels and several other clinical parameters were recorded. The effect of NS1 treatment of cultured fibroblasts and endothelial cells on the expressions of hyaluronan synthetic and catabolic enzymes and the hyaluronan receptor CD44, were determined, as have the effects on the formation of hyaluronan-rich matrices and endothelial permeability. FINDINGS Elevated serum hyaluronan levels (≥70 ng/ml) during early infection was found to be an independent predictor for occurrence of warning signs, and thus severe dengue fever. High circulating levels of the viral protein NS1, indicative of disease severity, correlated with high concentrations of serum hyaluronan. NS1 exposure decreased the expression of CD44 in differentiating endothelial cells impairing the integrity of vessel-like structures, and promoted the synthesis of hyaluronan in dermal fibroblasts and endothelial cells in synergy with dengue-induced pro-inflammatory mediators. Deposited hyaluronan-rich matrices around cells cultured in vitro recruited CD44-expressing macrophage-like cells, suggesting a mechanism for enhancement of inflammation. In cultured endothelial cells, perturbed hyaluronan-CD44 interactions enhanced endothelial permeability through modulation of VE-cadherin and cytoskeleton re-organization, and exacerbated the NS1-induced disruption of endothelial integrity. INTERPRETATION Pharmacological targeting of hyaluronan biosynthesis and/or its CD44-mediated signaling may limit the life-threatening vascular leakiness during moderate-to-severe dengue virus infection. FUND: This work was supported in part by grants from the Swedish Cancer Society (2018/337; 2016/445), the Swedish Research Council (2015-02757), the Ludwig Institute for Cancer Research, Uppsala University, the Ministry of Science and Technology, Taiwan (106-2314-B-037-088- and 106-2915-I-037-501-), Kaohsiung Medical University Hospital (KMUH103-3 T05) and Academy of Finland. The funders played no role in the design, interpretation or writing of the manuscript.
Collapse
Affiliation(s)
- Chun-Yu Lin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Box 582, SE-751 23 Uppsala, Sweden; Division of Infectious Diseases, Department of Internal Medicine, Infection Control Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; School of Medicine, Graduate Institute of Medicine, Sepsis Research Center, Center of Dengue Fever Control and Research, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Surgical Sciences, Uppsala University, Akademiska Hospital, 751 85 Uppsala, Sweden
| | - Constantinos Kolliopoulos
- Department of Medical Biochemistry and Microbiology, Uppsala University, Box 582, SE-751 23 Uppsala, Sweden
| | - Chung-Hao Huang
- Division of Infectious Diseases, Department of Internal Medicine, Infection Control Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; School of Medicine, Graduate Institute of Medicine, Sepsis Research Center, Center of Dengue Fever Control and Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jyrki Tenhunen
- Department of Surgical Sciences, Uppsala University, Akademiska Hospital, 751 85 Uppsala, Sweden; Critical Care Medicine Research Group, Department of Intensive Care, Tampere University Hospital, Tampere, Finland
| | - Carl-Henrik Heldin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Box 582, SE-751 23 Uppsala, Sweden
| | - Yen-Hsu Chen
- School of Medicine, Graduate Institute of Medicine, Sepsis Research Center, Center of Dengue Fever Control and Research, Kaohsiung Medical University, Kaohsiung, Taiwan; Deparent of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan; Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, HsinChu, Taiwan.
| | - Paraskevi Heldin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Box 582, SE-751 23 Uppsala, Sweden.
| |
Collapse
|
2031
|
Co-immobilization of ACH 11 antithrombotic peptide and CAG cell-adhesive peptide onto vascular grafts for improved hemocompatibility and endothelialization. Acta Biomater 2019; 97:344-359. [PMID: 31377424 DOI: 10.1016/j.actbio.2019.07.057] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 07/28/2019] [Accepted: 07/30/2019] [Indexed: 11/20/2022]
Abstract
Surface modification by conjugating biomolecules has been widely proved to enhance biocompatibility of small-caliber artificial vascular grafts. In this study, we aimed at developing a multifunctional vascular graft that provides not only good hemocompatibility but also in situ rapid endothelialization. Herein, a vascular graft (inner diameter ∼2 mm) was fabricated by electrospinning with poly(lactic acid-co-caprolactone) and gelatin, and then biofunctionalized with antithrombotic peptide with sequence LTFPRIVFVLG (ACH11) and cell adhesion peptide with sequence CAG through adhesive poly(dopamine) coating. We developed this graft with the synergistic properties of low thrombogenicity and rapid endothelialization. The successful grafting of both CAG and ACH11 peptides was confirmed by Fourier transform infrared spectroscopy and X-ray photoelectron spectroscopy. The surface micromorphology of the modified surfaces was observed by field emission scanning electron microscopy. Our results demonstrated that the multifunctional surface suppressed the denaturation of absorbed fibrinogen, hindered coagulation factor Xa activation, and inhibited platelet adhesion and aggregation. Importantly, this modified surface could selectively enhance endothelial cells adhesion, proliferation and release of nitric oxide. Upon in vivo implantation of 6 weeks, the multifunctional vascular graft showed improved patency and superior vascular endothelialization. Overall, the results effectively demonstrated that the co-immobilization of ACH11 and CAG provided a promising method for the improvement of hemocompatibility and endothelialization of vascular grafts. STATEMENT OF SIGNIFICANCE: Electrospun small-caliber vascular grafts are increasingly used to treat cardiovascular diseases. Despite their success related to their good biodegradation and mechanical strength, they have some drawbacks, such as low hemocompatibility and endothelialization. The single-function ligands are insufficient to modify surface with both good hemocompatibility and rapid endothelialization simultaneously. Therefore, we functionalized electrospun vascular graft by novel antithrombotic peptide and cell-adhesive peptide to construct superior anticoagulation and ECs-selective adhesion surface in present study. The multifunctional vascular grafts benefit for high long-term patency and rapid endothelialization.
Collapse
|
2032
|
Park S, Ranjbarvaziri S, Lay FD, Zhao P, Miller MJ, Dhaliwal JS, Huertas-Vazquez A, Wu X, Qiao R, Soffer JM, Rau C, Wang Y, Mikkola HKA, Lusis AJ, Ardehali R. Genetic Regulation of Fibroblast Activation and Proliferation in Cardiac Fibrosis. Circulation 2019; 138:1224-1235. [PMID: 29950403 DOI: 10.1161/circulationaha.118.035420] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Genetic diversity and the heterogeneous nature of cardiac fibroblasts (CFbs) have hindered characterization of the molecular mechanisms that regulate cardiac fibrosis. The Hybrid Mouse Diversity Panel offers a valuable tool to examine genetically diverse cardiac fibroblasts and their role in fibrosis. METHODS Three strains of mice (C57BL/6J, C3H/HeJ, and KK/HlJ) were selected from the Hybrid Mouse Diversity Panel and treated with either isoproterenol (ISO) or saline by an intraperitoneally implanted osmotic pump. After 21 days, cardiac function and levels of fibrosis were measured by echocardiography and trichrome staining, respectively. Activation and proliferation of CFbs were measured by in vitro and in vivo assays under normal and injury conditions. RNA sequencing was done on isolated CFbs from each strain. Results were analyzed by Ingenuity Pathway Analysis and validated by reverse transcription-qPCR, immunohistochemistry, and ELISA. RESULTS ISO treatment in C57BL/6J, C3H/HeJ, and KK/HlJ mice resulted in minimal, moderate, and extensive levels of fibrosis, respectively (n=7-8 hearts per condition). Isolated CFbs treated with ISO exhibited strain-specific increases in the levels of activation but showed comparable levels of proliferation. Similar results were found in vivo, with fibroblast activation, and not proliferation, correlating with the differential levels of cardiac fibrosis after ISO treatment. RNA sequencing revealed that CFbs from each strain exhibit unique gene expression changes in response to ISO. We identified Ltbp2 as a commonly upregulated gene after ISO treatment. Expression of LTBP2 was elevated and specifically localized in the fibrotic regions of the myocardium after injury in mice and in human heart failure patients. CONCLUSIONS This study highlights the importance of genetic variation in cardiac fibrosis by using multiple inbred mouse strains to characterize CFbs and their response to ISO treatment. Our data suggest that, although fibroblast activation is a response that parallels the extent of scar formation, proliferation may not necessarily correlate with levels of fibrosis. In addition, by comparing CFbs from multiple strains, we identified pathways as potential therapeutic targets and LTBP2 as a marker for fibrosis, with relevance to patients with underlying myocardial fibrosis.
Collapse
Affiliation(s)
- Shuin Park
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine (S.P., S.R., P.Z., M.J.M., J.S.D., A.H.-V., X.W., R.Q., J.M.S., A.J.L., R.A.), University of California, Los Angeles.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research (S.P., S.R., F.D.L., P.Z., M.J.M., J.S.D., R.Q., J.M.S., H.K.A.M., R.A.), University of California, Los Angeles.,Molecular, Cellular, and Integrative Physiology Graduate Program (S.P., S.R., R.A.), University of California, Los Angeles
| | - Sara Ranjbarvaziri
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine (S.P., S.R., P.Z., M.J.M., J.S.D., A.H.-V., X.W., R.Q., J.M.S., A.J.L., R.A.), University of California, Los Angeles.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research (S.P., S.R., F.D.L., P.Z., M.J.M., J.S.D., R.Q., J.M.S., H.K.A.M., R.A.), University of California, Los Angeles.,Molecular, Cellular, and Integrative Physiology Graduate Program (S.P., S.R., R.A.), University of California, Los Angeles
| | - Fides D Lay
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine (S.P., S.R., P.Z., M.J.M., J.S.D., A.H.-V., X.W., R.Q., J.M.S., A.J.L., R.A.), University of California, Los Angeles.,Department of Molecular, Cell, and Developmental Biology (F.D.L., H.K.A.M.), University of California, Los Angeles
| | - Peng Zhao
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine (S.P., S.R., P.Z., M.J.M., J.S.D., A.H.-V., X.W., R.Q., J.M.S., A.J.L., R.A.), University of California, Los Angeles.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research (S.P., S.R., F.D.L., P.Z., M.J.M., J.S.D., R.Q., J.M.S., H.K.A.M., R.A.), University of California, Los Angeles
| | - Mark J Miller
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine (S.P., S.R., P.Z., M.J.M., J.S.D., A.H.-V., X.W., R.Q., J.M.S., A.J.L., R.A.), University of California, Los Angeles.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research (S.P., S.R., F.D.L., P.Z., M.J.M., J.S.D., R.Q., J.M.S., H.K.A.M., R.A.), University of California, Los Angeles
| | - Jasmeet S Dhaliwal
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine (S.P., S.R., P.Z., M.J.M., J.S.D., A.H.-V., X.W., R.Q., J.M.S., A.J.L., R.A.), University of California, Los Angeles.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research (S.P., S.R., F.D.L., P.Z., M.J.M., J.S.D., R.Q., J.M.S., H.K.A.M., R.A.), University of California, Los Angeles
| | - Adriana Huertas-Vazquez
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine (S.P., S.R., P.Z., M.J.M., J.S.D., A.H.-V., X.W., R.Q., J.M.S., A.J.L., R.A.), University of California, Los Angeles
| | - Xiuju Wu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine (S.P., S.R., P.Z., M.J.M., J.S.D., A.H.-V., X.W., R.Q., J.M.S., A.J.L., R.A.), University of California, Los Angeles
| | - Rong Qiao
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine (S.P., S.R., P.Z., M.J.M., J.S.D., A.H.-V., X.W., R.Q., J.M.S., A.J.L., R.A.), University of California, Los Angeles.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research (S.P., S.R., F.D.L., P.Z., M.J.M., J.S.D., R.Q., J.M.S., H.K.A.M., R.A.), University of California, Los Angeles
| | - Justin M Soffer
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine (S.P., S.R., P.Z., M.J.M., J.S.D., A.H.-V., X.W., R.Q., J.M.S., A.J.L., R.A.), University of California, Los Angeles.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research (S.P., S.R., F.D.L., P.Z., M.J.M., J.S.D., R.Q., J.M.S., H.K.A.M., R.A.), University of California, Los Angeles
| | - Christoph Rau
- Anesthesiology and Perioperative Medicine (C.R., Y.W.), University of California, Los Angeles
| | - Yibin Wang
- Anesthesiology and Perioperative Medicine (C.R., Y.W.), University of California, Los Angeles
| | - Hanna K A Mikkola
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research (S.P., S.R., F.D.L., P.Z., M.J.M., J.S.D., R.Q., J.M.S., H.K.A.M., R.A.), University of California, Los Angeles.,Department of Molecular, Cell, and Developmental Biology (F.D.L., H.K.A.M.), University of California, Los Angeles.,Molecular Biology Institute (H.K.A.M., A.J.L., R.A.), University of California, Los Angeles
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine (S.P., S.R., P.Z., M.J.M., J.S.D., A.H.-V., X.W., R.Q., J.M.S., A.J.L., R.A.), University of California, Los Angeles.,Department of Microbiology, Immunology, and Molecular Genetics (A.J.L.), University of California, Los Angeles.,Molecular Biology Institute (H.K.A.M., A.J.L., R.A.), University of California, Los Angeles.,Department of Human Genetics (A.J.L.), University of California, Los Angeles
| | - Reza Ardehali
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine (S.P., S.R., P.Z., M.J.M., J.S.D., A.H.-V., X.W., R.Q., J.M.S., A.J.L., R.A.), University of California, Los Angeles.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research (S.P., S.R., F.D.L., P.Z., M.J.M., J.S.D., R.Q., J.M.S., H.K.A.M., R.A.), University of California, Los Angeles.,Molecular, Cellular, and Integrative Physiology Graduate Program (S.P., S.R., R.A.), University of California, Los Angeles.,Molecular Biology Institute (H.K.A.M., A.J.L., R.A.), University of California, Los Angeles
| |
Collapse
|
2033
|
Wang M, Wang C, Chen M, Xi Y, Cheng W, Mao C, Xu T, Zhang X, Lin C, Gao W, Guo Y, Lei B. Efficient Angiogenesis-Based Diabetic Wound Healing/Skin Reconstruction through Bioactive Antibacterial Adhesive Ultraviolet Shielding Nanodressing with Exosome Release. ACS NANO 2019; 13:10279-10293. [PMID: 31483606 DOI: 10.1021/acsnano.9b03656] [Citation(s) in RCA: 359] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Diabetic wound healing and angiogenesis remain a worldwide challenge for both clinic and research. The use of adipose stromal cell derived exosomes delivered by bioactive dressing provides a potential strategy for repairing diabetic wounds with less scar formation and fast healing. In this study, we fabricated an injectable adhesive thermosensitive multifunctional polysaccharide-based dressing (FEP) with sustained pH-responsive exosome release for promoting angiogenesis and diabetic wound healing. The FEP dressing possessed multifunctional properties including efficient antibacterial activity/multidrug-resistant bacteria, fast hemostatic ability, self-healing behavior, and tissue-adhesive and good UV-shielding performance. FEP@exosomes (FEP@exo) can significantly enhance the proliferation, migration, and tube formation of endothelial cells in vitro. In vivo results from a diabetic full-thickness cutaneous wound model showed that FEP@exo dressing accelerated the wound healing by stimulating the angiogenesis process of the wound tissue. The enhanced cell proliferation, granulation tissue formation, collagen deposition, remodeling, and re-epithelialization probably lead to the fast healing with less scar tissue formation and skin appendage regeneration. This study showed that combining bioactive molecules into multifunctional dressing should have great potential in achieving satisfactory healing in diabetic and other vascular-impaired related wounds.
Collapse
Affiliation(s)
- Min Wang
- Frontier Institute of Science and Technology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology , Xi'an Jiaotong University , Xi'an 710000 , China
| | - Chenggui Wang
- Key Laboratory of Orthopedics of Zhejiang Province , Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou 325027 , China
- Department of Orthopedics Surgery , Second Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou 310009 , China
| | - Mi Chen
- Frontier Institute of Science and Technology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology , Xi'an Jiaotong University , Xi'an 710000 , China
| | - Yuewei Xi
- Frontier Institute of Science and Technology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology , Xi'an Jiaotong University , Xi'an 710000 , China
| | - Wei Cheng
- Frontier Institute of Science and Technology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology , Xi'an Jiaotong University , Xi'an 710000 , China
| | - Cong Mao
- Key Laboratory of Orthopedics of Zhejiang Province , Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou 325027 , China
| | - Tianzhen Xu
- Key Laboratory of Orthopedics of Zhejiang Province , Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou 325027 , China
| | - Xingxing Zhang
- Department of Burn , First Affiliated Hospital of Wenzhou Medical University , Wenzhou 325000 , China
| | - Cai Lin
- Department of Burn , First Affiliated Hospital of Wenzhou Medical University , Wenzhou 325000 , China
| | - Weiyang Gao
- Key Laboratory of Orthopedics of Zhejiang Province , Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou 325027 , China
| | - Yi Guo
- Department of Biologic and Materials Science , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Bo Lei
- Frontier Institute of Science and Technology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology , Xi'an Jiaotong University , Xi'an 710000 , China
| |
Collapse
|
2034
|
Lou Q, Li Y, Hou B, Liu Y, Zhang Y, Hao J, Ma Y. Heat shock transcription factor 1 affects kidney tubular cell migration by regulating the TGF‑β1‑Smad2/3 signaling pathway. Mol Med Rep 2019; 20:4323-4330. [PMID: 31545442 DOI: 10.3892/mmr.2019.10689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 08/23/2019] [Indexed: 11/06/2022] Open
Abstract
Cell migration is important for renal recovery from tubular cell injury. Heat shock transcription factor 1 (HSF1) is a well‑studied regulatory factor that is active during acute kidney injury. HSF1 is also involved in the migration process during tumor metastasis. Therefore, we hypothesized that HSF1 may promote the recovery of renal function by affecting kidney tubular cell migration. A wound healing assay was used to examine the cell migration rate. The results demonstrated that the migration of rat kidney proximal tubular cells (RPTCs) was increased following knockdown of HSF1. In addition, the invasion ability of HSF1 knockdown RPTCs was also significantly upregulated. The present study also identified that transforming growth factor‑β1 (TGF‑β1) was highly expressed at the edge of the wound in control cells, and its expression was further increased upon knockdown of HSF1. Inhibition of TGF‑β1 signaling prevented RPTC HSF1 knockdown cell migration, suggesting that HSF1‑regulated RPTC cell migration was dependent on the TGF‑β1 signaling pathway. Furthermore, phosphorylation of TGF‑β1 and Smad2/3 was induced in HSF1 knockdown cells. Together, these results suggest that HSF1 may suppress RPTC migration by inhibiting the activation of the TGF‑β1‑Smad2/3 signaling pathway.
Collapse
Affiliation(s)
- Qiang Lou
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Yuanyuan Li
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Beibei Hou
- International Office of Henan University, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Yonglian Liu
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Yan Zhang
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Jielu Hao
- Department of Nephrology, Changzheng Hospital Affiliated to Second Military Medical University, Shanghai 200003, P.R. China
| | - Yuanfang Ma
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, Henan 475004, P.R. China
| |
Collapse
|
2035
|
Pandolfi L, Frangipane V, Bocca C, Marengo A, Tarro Genta E, Bozzini S, Morosini M, D'Amato M, Vitulo S, Monti M, Comolli G, Scupoli MT, Fattal E, Arpicco S, Meloni F. Hyaluronic Acid-Decorated Liposomes as Innovative Targeted Delivery System for Lung Fibrotic Cells. Molecules 2019; 24:molecules24183291. [PMID: 31509965 PMCID: PMC6766933 DOI: 10.3390/molecules24183291] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/05/2019] [Accepted: 09/07/2019] [Indexed: 12/19/2022] Open
Abstract
Collagen Tissue Disease-associated Interstitial Lung Fibrosis (CTD-ILDs) and Bronchiolitis Obliterans Syndrome (BOS) represent severe lung fibrogenic disorders, characterized by fibro-proliferation with uncontrolled extracellular matrix deposition. Hyaluronic acid (HA) plays a key role in fibrosis with its specific receptor, CD44, overexpressed by CTD-ILD and BOS cells. The aim is to use HA-liposomes to develop an inhalatory treatment for these diseases. Liposomes with HA of two molecular weights were prepared and characterized. Targeting efficiency was assessed toward CTD-ILD and BOS cells by flow cytometry and confocal microscopy and immune modulation by RT-PCR and ELISA techniques. HA-liposomes were internalized by CTD-ILD and BOS cells expressing CD44, and this effect increased with higher HA MW. In THP-1 cells, HA-liposomes decreased pro-inflammatory cytokines IL-1β, IL-12, and anti-fibrotic VEGF transcripts but increased TGF-β mRNA. However, upon analyzing TGF-β release from healthy donors-derived monocytes, we found liposomes did not alter the release of active pro-fibrotic cytokine. All liposomes induced mild activation of neutrophils regardless of the presence of HA. HA liposomes could be also applied for lung fibrotic diseases, being endowed with low pro-inflammatory activity, and results confirmed that higher MW HA are associated to an increased targeting efficiency for CD44 expressing LFs-derived from BOS and CTD-ILD patients.
Collapse
Affiliation(s)
- Laura Pandolfi
- Research Laboratory of Lung Diseases, Section of Cell Biology, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy.
| | - Vanessa Frangipane
- Research Laboratory of Lung Diseases, Section of Cell Biology, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy.
| | - Claudia Bocca
- Department of Clinical and Biological Sciences, University of Turin, 10125 Turin, Italy.
| | - Alessandro Marengo
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy.
| | - Erika Tarro Genta
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy.
| | - Sara Bozzini
- Research Laboratory of Lung Diseases, Section of Cell Biology, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy.
| | - Monica Morosini
- Research Laboratory of Lung Diseases, Section of Cell Biology, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy.
| | - Maura D'Amato
- Research Laboratory of Lung Diseases, Section of Cell Biology, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy.
| | - Simone Vitulo
- Research Laboratory of Lung Diseases, Section of Cell Biology, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy.
| | - Manuela Monti
- Laboratory of Biotechnology, Center of Regenerative Medicine Research, IRCCS San Matteo Foundation, 27100 Pavia, Italy.
| | - Giuditta Comolli
- Experimental Research Laboratories, Biotechnology Area, IRCCS San Matteo Foundation, 27100 Pavia, Italy.
- Molecular Virology Unit, Microbiology and Virology Department, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy.
| | - Maria Teresa Scupoli
- Research Center LURM, Interdepartmental Laboratory of Medical Research, University of Verona, 37134 Verona, Italy.
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37129 Verona, Italy.
| | - Elias Fattal
- Institut Galien Paris-Sud, CNRS, Université Paris-Sud, Université Paris-Saclay, 922996 Châtenay-Malabry, France.
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy.
| | - Federica Meloni
- Research Laboratory of Lung Diseases, Section of Cell Biology, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy.
- Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy.
| |
Collapse
|
2036
|
Zeng X, Li C, Li Y, Yu H, Fu P, Hong HG, Zhang W. A network-based variable selection approach for identification of modules and biomarker genes associated with end-stage kidney disease. Nephrology (Carlton) 2019; 25:775-784. [PMID: 31464346 DOI: 10.1111/nep.13655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2019] [Indexed: 02/05/2023]
Abstract
AIMS Intervention for end-stage kidney disease (ESKD), which is associated with adverse prognoses and major economic burdens, is challenging due to its complex pathogenesis. The study was performed to identify biomarker genes and molecular mechanisms for ESKD by bioinformatics approach. METHODS Using the Gene Expression Omnibus dataset GSE37171, this study identified pathways and genomic biomarkers associated with ESKD via a multi-stage knowledge discovery process, including identification of modules of genes by weighted gene co-expression network analysis, discovery of important involved pathways by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses, selection of differentially expressed genes by the empirical Bayes method, and screening biomarker genes by the least absolute shrinkage and selection operator (Lasso) logistic regression. The results were validated using GSE70528, an independent testing dataset. RESULTS Three clinically important gene modules associated with ESKD, were identified by weighted gene co-expression network analysis. Within these modules, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses revealed important biological pathways involved in ESKD, including transforming growth factor-β and Wnt signalling, RNA-splicing, autophagy and chromatin and histone modification. Furthermore, Lasso logistic regression was conducted to identify five final genes, namely, CNOT8, MST4, PPP2CB, PCSK7 and RBBP4 that are differentially expressed and associated with ESKD. The accuracy of the final model in distinguishing the ESKD cases and controls was 96.8% and 91.7% in the training and validation datasets, respectively. CONCLUSION Network-based variable selection approaches can identify biological pathways and biomarker genes associated with ESKD. The findings may inform more in-depth follow-up research and effective therapy.
Collapse
Affiliation(s)
- Xiaoxi Zeng
- West China Biomedical Big Data Center, West China School of Medicine (West China Hospital), Sichuan University, Chengdu, China.,Division of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China.,Medical Big Data Center, Sichuan University, Chengdu, China
| | - Chunyang Li
- West China Biomedical Big Data Center, West China School of Medicine (West China Hospital), Sichuan University, Chengdu, China.,Medical Big Data Center, Sichuan University, Chengdu, China
| | - Yi Li
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Haopeng Yu
- West China Biomedical Big Data Center, West China School of Medicine (West China Hospital), Sichuan University, Chengdu, China.,Medical Big Data Center, Sichuan University, Chengdu, China
| | - Ping Fu
- Division of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China.,Medical Big Data Center, Sichuan University, Chengdu, China
| | - Hyokyoung G Hong
- Department of Statistics and Probability, Michigan State University, East Lansing, Michigan, USA
| | - Wei Zhang
- West China Biomedical Big Data Center, West China School of Medicine (West China Hospital), Sichuan University, Chengdu, China.,Medical Big Data Center, Sichuan University, Chengdu, China
| |
Collapse
|
2037
|
Scavenius C, Poulsen EC, Thøgersen IB, Roebuck M, Frostick S, Bou-Gharios G, Yamamoto K, Deleuran B, Enghild JJ. Matrix-degrading protease ADAMTS-5 cleaves inter-α-inhibitor and releases active heavy chain 2 in synovial fluids from arthritic patients. J Biol Chem 2019; 294:15495-15504. [PMID: 31484722 DOI: 10.1074/jbc.ra119.008844] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/29/2019] [Indexed: 12/20/2022] Open
Abstract
Destruction of the cartilage matrix in joints is an important feature of arthritis. Proteolytic degradation of cartilage glycoproteins can contribute to the loss of matrix integrity. Human inter-α-inhibitor (IαI), which stabilizes the extracellular matrix, is composed of the light-chain serine proteinase inhibitor bikunin and two homologous heavy chains (HC1 and HC2) covalently linked through chondroitin 4-sulfate. Inflammation promotes the transfer of HCs from chondroitin 4-sulfate to hyaluronan by tumor necrosis factor-stimulated gene-6 protein (TSG-6). This reaction generates a covalent complex between the heavy chains and hyaluronan that can promote leukocyte invasion. This study demonstrates that both IαI and the HC-hyaluronan complex are substrates for the extracellular matrix proteases ADAMTS-5 and matrix metalloprotease (MMP) -3, -7, and -13. The major cleavage sites for all four proteases are found in the C terminus of HC2. ADAMTS-5 and MMP-7 displayed the highest activity toward HC2. ADAMTS-5 degradation products were identified in mass spectrometric analysis of 29 of 33 arthropathic patients, indicating that ADAMTS-5 cleavage occurs in synovial fluid in arthritis. After cleavage, free HC2, together with TSG-6, is able to catalyze the transfer of heavy chains to hyaluronan. The release of extracellular matrix bound HC2 is likely to increase the mobility of the HC2/TSG-6 catalytic unit and consequently increase the rate of the HC transfer reaction. Ultimately, ADAMTS-5 cleavage of HC2 could alter the physiological and mechanical properties of the extracellular matrix and contribute to the progression of arthritis.
Collapse
Affiliation(s)
- Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | | | - Ida B Thøgersen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Margaret Roebuck
- Department of Molecular and Clinical Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Simon Frostick
- Department of Molecular and Clinical Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - George Bou-Gharios
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Kazuhiro Yamamoto
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Bent Deleuran
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark.,Department of Rheumatology, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
2038
|
Seow BKL, McDougall ARA, Short KL, Wallace MJ, Hooper SB, Cole TJ. Identification of Betamethasone-Regulated Target Genes and Cell Pathways in Fetal Rat Lung Mesenchymal Fibroblasts. Endocrinology 2019; 160:1868-1884. [PMID: 31107524 DOI: 10.1210/en.2018-01071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 05/14/2019] [Indexed: 02/07/2023]
Abstract
Preterm birth is characterized by severe lung immaturity that is frequently treated antenatally or postnatally with the synthetic steroid betamethasone. The underlying cellular targets and pathways stimulated by betamethasone in the fetal lung are poorly defined. In this study, betamethasone was compared with corticosterone in steroid-treated primary cultures of fetal rat lung fibroblasts stimulated for 6 hours and analyzed by whole-cell transcriptome sequencing and glucocorticoid (GC) receptor (GR) chromatin immunoprecipitation sequencing (ChIP-Seq) analysis. Strikingly, betamethasone stimulated a much stronger transcriptional response compared with corticosterone for both induced and repressed genes. A total of 483 genes were significantly stimulated by betamethasone or corticosterone, with 476 stimulated by both steroids, indicating a strong overlap in regulation. Changes in mRNA levels were confirmed by quantitative PCR for eight induced and repressed target genes. Pathway analysis identified cell proliferation and cytoskeletal/cell matrix remodeling pathways as key processes regulated by both steroids. One target, transglutaminase 2 (Tgm2), was localized to fetal lung mesenchymal cells. Tgm2 mRNA and protein levels were strongly increased in fibroblasts by both steroids. Whole-genome GR ChIP-Seq analysis with betamethasone identified GC response element-binding sites close to the previously characterized GR target genes Per1, Dusp1, Fkbp5, and Sgk1 and near the genes identified by transcriptome sequencing encoding Crispld2, Tgm2, Hif3α, and Kdr, defining direct genomic induction of expression in fetal lung fibroblasts via the GR. These results demonstrate that betamethasone stimulates specific genes and cellular pathways controlling cell proliferation and extracellular matrix remodeling in lung mesenchymal fibroblasts, providing a basis for betamethasone's treatment efficacy in preterm birth.
Collapse
Affiliation(s)
- Bennet K L Seow
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Annie R A McDougall
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Kelly L Short
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - Megan J Wallace
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Stuart B Hooper
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Timothy J Cole
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
- Division of Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| |
Collapse
|
2039
|
IDH2 deficiency impairs cutaneous wound healing via ROS-dependent apoptosis. Biochim Biophys Acta Mol Basis Dis 2019; 1865:165523. [PMID: 31376482 DOI: 10.1016/j.bbadis.2019.07.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 01/21/2023]
Abstract
Dermal fibroblasts are mesenchymal cells found between the skin epidermis and subcutaneous tissue that play a pivotal role in cutaneous wound healing by synthesizing fibronectin (a component of the extracellular matrix), secreting angiogenesis factors, and generating strong contractile forces. In wound healing, low concentrations of reactive oxygen species (ROS) are essential in combating invading microorganisms and in cell-survival signaling. However, excessive ROS production impairs fibroblasts. Mitochondrial NADP+-dependent isocitrate dehydrogenase (IDH2) is a key enzyme that regulates the mitochondrial redox balance and reduces oxidative stress-induced cell injury through the generation of NADPH. In the present study, the downregulation of IDH2 expression resulted in an increase in cell apoptosis in mouse skin through ROS-dependent ATM-mediated p53 signaling. IDH2 deficiency also delayed cutaneous wound healing in mice and impaired dermal fibroblast function. Furthermore, pretreatment with the mitochondria-targeted antioxidant mito-TEMPO alleviated the apoptosis induced by IDH2 deficiency both in vitro and in vivo. Together, our findings highlight the role of IDH2 in cutaneous wound healing in association with mitochondrial ROS.
Collapse
|
2040
|
Sarkar A, Chanda A, Regmi SC, Karve K, Deng L, Jay GD, Jirik FR, Schmidt TA, Bonni S. Recombinant human PRG4 (rhPRG4) suppresses breast cancer cell invasion by inhibiting TGFβ-Hyaluronan-CD44 signalling pathway. PLoS One 2019; 14:e0219697. [PMID: 31361756 PMCID: PMC6667139 DOI: 10.1371/journal.pone.0219697] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/28/2019] [Indexed: 01/02/2023] Open
Abstract
Metastasis is the major cause of cancer-related morbidity and mortality. The ability of cancer cells to become invasive and migratory contribute significantly to metastatic growth, which necessitates the identification of novel anti-migratory and anti-invasive therapeutic approaches. Proteoglycan 4 (PRG4), a mucin-like glycoprotein, contributes to joint synovial homeostasis through its friction-reducing and anti-adhesive properties. Adhesion to surrounding extracellular matrix (ECM) components is critical for cancer cells to invade the ECM and eventually become metastatic, raising the question whether PRG4 has an anti-invasive effect on cancer cells. Here, we report that a full-length recombinant human PRG4 (rhPRG4) suppresses the ability of the secreted protein transforming growth factor beta (TGFβ) to induce phenotypic disruption of three-dimensional human breast cancer cell-derived organoids by reducing ligand-induced cell invasion. In mechanistic studies, we find that rhPRG4 suppresses TGFβ-induced invasiveness of cancer cells by inhibiting the downstream hyaluronan (HA)-cell surface cluster of differentiation 44 (CD44) signalling axis. Furthermore, we find that rhPRG4 represses TGFβ-dependent increase in the protein abundance of CD44 and of the enzyme HAS2, which is involved in HA biosynthesis. It is widely accepted that TGFβ has both tumor suppressing and tumor promoting roles in cancer. The novel finding that rhPRG4 opposes HAS2 and CD44 induction by TGFβ has implications for downregulating the tumor promoting roles, while maintaining the tumor suppressive aspects of TGFβ actions. Finally, these findings point to rhPRG4's potential clinical utility as a therapeutic treatment for invasive and metastatic breast cancer.
Collapse
Affiliation(s)
- Anusi Sarkar
- The Arnie Charbonneau Cancer Institute and Department of Biochemistry & Molecular Biology, The Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Ayan Chanda
- The Arnie Charbonneau Cancer Institute and Department of Biochemistry & Molecular Biology, The Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Suresh C. Regmi
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Kunal Karve
- The Arnie Charbonneau Cancer Institute and Department of Biochemistry & Molecular Biology, The Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Lili Deng
- The Arnie Charbonneau Cancer Institute and Department of Biochemistry & Molecular Biology, The Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gregory D. Jay
- Department of Emergency Medicine—Alpert Medical School & School of Engineering, Brown University, Providence, Rhode Island, United States of America
| | - Frank R. Jirik
- The Arnie Charbonneau Cancer Institute and Department of Biochemistry & Molecular Biology, The Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Tannin A. Schmidt
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Biomedical Engineering Department, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- * E-mail: (SB); (TS)
| | - Shirin Bonni
- The Arnie Charbonneau Cancer Institute and Department of Biochemistry & Molecular Biology, The Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- * E-mail: (SB); (TS)
| |
Collapse
|
2041
|
Abstract
Tissue-engineered vascular grafts (TEVGs) are considered one of the most effective means of fabricating vascular grafts. However, for small-diameter TEVGs, there are ongoing issues regarding long-term patency and limitations related to long-term in vitro culture and immune reactions. The use of acellular TEVG is a more convincing method, which can achieve in situ blood vessel regeneration and better meet clinical needs. This review focuses on the current state of acellular TEVGs based on scaffolds and gives a summary of the methodologies and in vitro/in vivo test results related to acellular TEVGs obtained in recent years. Various strategies for improving the properties of acellular TEVGs are also discussed.
Collapse
|
2042
|
Ferroni L, Gardin C, Dalla Paola L, Campo G, Cimaglia P, Bellin G, Pinton P, Zavan B. Characterization of Dermal Stem Cells of Diabetic Patients. Cells 2019; 8:cells8070729. [PMID: 31315286 PMCID: PMC6678145 DOI: 10.3390/cells8070729] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 12/26/2022] Open
Abstract
Diabetic foot ulcers (DFUs) are lesions that involve loss of epithelium and dermis, sometimes involving deep structures, compartments, and bones. The aim of this work is to investigate the innate regenerative properties of dermal tissue around ulcers by the identification and analysis of resident dermal stem cells (DSCs). Dermal samples were taken at the edge of DFUs, and genes related to the wound healing process were analyzed by the real-time PCR array. The DSCs were isolated and analyzed by immunofluorescence, flow cytometry, and real-time PCR array to define their stemness properties. The gene expression profile of dermal tissue showed a dysregulation in growth factors, metalloproteinases, collagens, and integrins involved in the wound healing process. In the basal condition, diabetic DSCs adhered on the culture plate with spindle-shaped fibroblast-like morphology. They were positive to the mesenchymal stem cells markers CD44, CD73, CD90, and CD105, but negative for the hematopoietic markers CD14, CD34, CD45, and HLA-DR. In diabetic DSCs, the transcription of genes related to self-renewal and cell division were equivalent to that in normal DSCs. However, the expression of CCNA2, CCND2, CDK1, ALDH1A1, and ABCG2 was downregulated compared with that of normal DSCs. These genes are also related to cell cycle progression and stem cell maintenance. Further investigation will improve the understanding of the molecular mechanisms by which these genes together govern cell proliferation, revealing new strategies useful for future treatment of DFUs.
Collapse
Affiliation(s)
- Letizia Ferroni
- Maria Cecilia Hospital, GVM Care & Research, 48,033 Cotignola (RA), Italy
- University of Ferrara, Department of Medical Sciences, via Fossato di Mortara 70, 44,121 Ferrara, Italy
| | - Chiara Gardin
- Maria Cecilia Hospital, GVM Care & Research, 48,033 Cotignola (RA), Italy
- University of Ferrara, Department of Medical Sciences, via Fossato di Mortara 70, 44,121 Ferrara, Italy
| | - Luca Dalla Paola
- Maria Cecilia Hospital, GVM Care & Research, 48,033 Cotignola (RA), Italy
| | - Gianluca Campo
- Maria Cecilia Hospital, GVM Care & Research, 48,033 Cotignola (RA), Italy
- University of Ferrara, Department of Medical Sciences, via Fossato di Mortara 70, 44,121 Ferrara, Italy
| | - Paolo Cimaglia
- Maria Cecilia Hospital, GVM Care & Research, 48,033 Cotignola (RA), Italy
| | - Gloria Bellin
- Maria Cecilia Hospital, GVM Care & Research, 48,033 Cotignola (RA), Italy
- University of Ferrara, Department of Medical Sciences, via Fossato di Mortara 70, 44,121 Ferrara, Italy
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care & Research, 48,033 Cotignola (RA), Italy
- Dept. of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44,121 Ferrara, Italy
| | - Barbara Zavan
- Maria Cecilia Hospital, GVM Care & Research, 48,033 Cotignola (RA), Italy.
- University of Ferrara, Department of Medical Sciences, via Fossato di Mortara 70, 44,121 Ferrara, Italy.
| |
Collapse
|
2043
|
Wang L, Wang D, Zhou Y, Zhang Y, Li Q, Shen C. Fabrication of open‐porous PCL/PLA tissue engineering scaffolds and the relationship of foaming process, morphology, and mechanical behavior. POLYM ADVAN TECHNOL 2019. [DOI: 10.1002/pat.4701] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Lixia Wang
- School of Mechanics and Engineering ScienceZhengzhou University Zhengzhou 450001 China
- National Center for International Research of Micro‐Nano Molding TechnologyZhengzhou University Zhengzhou 450001 China
| | - Dongfang Wang
- School of Mechanics and Engineering ScienceZhengzhou University Zhengzhou 450001 China
- National Center for International Research of Micro‐Nano Molding TechnologyZhengzhou University Zhengzhou 450001 China
| | - Yiping Zhou
- School of Mechanics and Engineering ScienceZhengzhou University Zhengzhou 450001 China
- National Center for International Research of Micro‐Nano Molding TechnologyZhengzhou University Zhengzhou 450001 China
| | - Yantao Zhang
- School of Mechanics and Engineering ScienceZhengzhou University Zhengzhou 450001 China
- National Center for International Research of Micro‐Nano Molding TechnologyZhengzhou University Zhengzhou 450001 China
| | - Qian Li
- School of Mechanics and Engineering ScienceZhengzhou University Zhengzhou 450001 China
- National Center for International Research of Micro‐Nano Molding TechnologyZhengzhou University Zhengzhou 450001 China
| | - Changyu Shen
- National Center for International Research of Micro‐Nano Molding TechnologyZhengzhou University Zhengzhou 450001 China
| |
Collapse
|
2044
|
Identification of a pro-angiogenic functional role for FSP1-positive fibroblast subtype in wound healing. Nat Commun 2019; 10:3027. [PMID: 31289275 PMCID: PMC6617456 DOI: 10.1038/s41467-019-10965-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 06/04/2019] [Indexed: 12/26/2022] Open
Abstract
Fibrosis accompanying wound healing can drive the failure of many different organs. Activated fibroblasts are the principal determinants of post-injury pathological fibrosis along with physiological repair, making them a difficult therapeutic target. Although activated fibroblasts are phenotypically heterogeneous, they are not recognized as distinct functional entities. Using mice that express GFP under the FSP1 or αSMA promoter, we characterized two non-overlapping fibroblast subtypes from mouse hearts after myocardial infarction. Here, we report the identification of FSP1-GFP+ cells as a non-pericyte, non-hematopoietic fibroblast subpopulation with a predominant pro-angiogenic role, characterized by in vitro phenotypic/cellular/ultrastructural studies and in vivo granulation tissue formation assays combined with transcriptomics and proteomics. This work identifies a fibroblast subtype that is functionally distinct from the pro-fibrotic αSMA-expressing myofibroblast subtype. Our study has the potential to shift our focus towards viewing fibroblasts as molecularly and functionally heterogeneous and provides a paradigm to approach treatment for organ fibrosis. Activated fibroblasts are key contributors to tissue repair after cardiac injury. Here, Saraswati et al. identify and characterize a subpopulation of FSP1-positive cardiac fibroblasts with proangiogenic properties in infarcted hearts.
Collapse
|
2045
|
Moon J, Yoon JY, Yang JH, Kwon HH, Min S, Suh DH. Atrophic acne scar: a process from altered metabolism of elastic fibres and collagen fibres based on transforming growth factor-β1 signalling. Br J Dermatol 2019; 181:1226-1237. [PMID: 30822364 DOI: 10.1111/bjd.17851] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Atrophic acne scar, a persistent sequela from acne, is undesirably troubling to many patients due to its cosmetic and psychosocial aspects. Although there have been some reports emphasizing the role of early inflammatory responses in atrophic acne scarring, evolving perspectives on the detailed pathogenic processes are promptly needed. OBJECTIVES Examining the histological, immunological and molecular changes in early acne lesions susceptible to atrophic scarring can provide new insights to understand the pathophysiology of atrophic acne scar. METHODS We experimentally validated several early fundamental hallmarks accounting for the transition of early acne lesions to atrophic scars by comparing molecular profiles of skin and acne lesions between patients who were prone to scar (APS) or not (ANS). RESULTS In APS, compared with ANS, devastating degradation of elastic fibres and collagen fibres occurred in the dermis, followed by their incomplete recovery. Abnormally excessive inflammation mediated by innate immunity with T helper 17 and T helper 1 cells was observed. Epidermal proliferation was significantly diminished. Transforming growth factor (TGF)-β1 was drastically elevated in APS, suggesting that aberrant TGF-β1 signalling is an underlying modulator of all of these pathological processes. CONCLUSIONS These results may provide a basis for understanding the pathogenesis of atrophic acne scarring. Reduction of excessive inflammation and TGF-β1 signalling in early acne lesions is expected to facilitate the protection of normal extracellular matrix metabolism and ultimately the prevention of atrophic scar formation. What's already known about this topic? The dermis of atrophic acne scars shows alteration of extracellular matrix components such as collagen fibres. Inflammation in acne lesions is associated with the development of acne scars. What does this study add? Abnormalities in the metabolism of collagen fibres and elastic fibres were observed in the early developmental stages of acne lesions that were progressing into atrophic scars. Exacerbated inflammation and aberrant epidermal proliferation by increased transforming growth factor (TGF)-β1 signalling may affect the abnormal extracellular matrix metabolism. What is the translational message? Abnormal changes in elastic fibres and collagen fibres are found in the early developmental process of acne in patients who are prone to atrophic scarring. An early treatment regimen strongly inhibiting inflammation and TGF-β1 signalling to help the normal recovery of the extracellular matrix components is required to prevent atrophic scarring.
Collapse
Affiliation(s)
- J Moon
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Acne, Rosacea, Seborrheic Dermatitis and Hidradenitis Suppurativa Research Laboratory, Seoul National University Hospital, Seoul, Republic of Korea
| | - J Y Yoon
- Acne, Rosacea, Seborrheic Dermatitis and Hidradenitis Suppurativa Research Laboratory, Seoul National University Hospital, Seoul, Republic of Korea
| | - J H Yang
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Acne, Rosacea, Seborrheic Dermatitis and Hidradenitis Suppurativa Research Laboratory, Seoul National University Hospital, Seoul, Republic of Korea
| | - H H Kwon
- Oaro Dermatology Clinic, Seoul, Republic of Korea
| | - S Min
- SnU Dermatology Clinic, Seoul, Republic of Korea
| | - D H Suh
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Acne, Rosacea, Seborrheic Dermatitis and Hidradenitis Suppurativa Research Laboratory, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
2046
|
Wang Y, Mack JA, Maytin EV. CD44 inhibits α-SMA gene expression via a novel G-actin/MRTF-mediated pathway that intersects with TGFβR/p38MAPK signaling in murine skin fibroblasts. J Biol Chem 2019; 294:12779-12794. [PMID: 31285260 DOI: 10.1074/jbc.ra119.007834] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/25/2019] [Indexed: 01/10/2023] Open
Abstract
Well-regulated differentiation of fibroblasts into myofibroblasts (MF) is critical for skin wound healing. Neoexpression of α-smooth muscle actin (α-SMA), an established marker for MF differentiation, is driven by TGFβ receptor (TGFβR)-mediated signaling. Hyaluronan (HA) and its receptor CD44 may also participate in this process. To further understand this process, primary mouse skin fibroblasts were isolated and treated in vitro with recombinant TGF-β1 (rTGF-β1) to induce α-SMA expression. CD44 expression was also increased. Paradoxically, CD44 knockdown by RNA interference (RNAi) led to increased α-SMA expression and α-SMA-containing stress fibers. Removal of extracellular HA or inhibition of HA synthesis had no effect on α-SMA levels, suggesting a dispensable role for HA. Exploration of mechanisms linking CD44 knockdown to α-SMA induction, using RNAi and chemical inhibitors, revealed a requirement for noncanonical TGFβR signaling through p38MAPK. Decreased monomeric G-actin but increased filamentous F-actin following CD44 RNAi suggested a possible role for myocardin-related transcription factor (MRTF), a known regulator of α-SMA transcription and itself regulated by G-actin binding. CD44 RNAi promoted nuclear accumulation of MRTF and the binding to its transcriptional cofactor SRF. MRTF knockdown abrogated the increased α-SMA expression caused by CD44 RNAi, suggesting that MRTF is required for CD44-mediated regulation of α-SMA. Finally, chemical inhibition of p38MAPK reversed nuclear MRTF accumulation after rTGF-β1 addition or CD44 RNAi, revealing a central involvement of p38MAPK in both cases. We concluded that CD44 regulates α-SMA gene expression through cooperation between two intersecting signaling pathways, one mediated by G-actin/MRTF and the other via TGFβR/p38MAPK.
Collapse
Affiliation(s)
- Yan Wang
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Judith A Mack
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195.,Department of Dermatology, Dermatology and Plastic Surgery Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Edward V Maytin
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195 .,Department of Dermatology, Dermatology and Plastic Surgery Institute, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|
2047
|
Genomic instability and innate immune responses to self-DNA in progeria. GeroScience 2019; 41:255-266. [PMID: 31280482 DOI: 10.1007/s11357-019-00082-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 06/21/2019] [Indexed: 12/15/2022] Open
Abstract
In the last decade, we have seen increasing evidence of the importance of structural nuclear proteins such as lamins in nuclear architecture and compartmentalization of genome function and in the maintenance of mechanical stability and genome integrity. With over 400 mutations identified in the LMNA gene (encoding for A-type lamins) associated with more than ten distinct degenerative disorders, the role of lamins as genome caretakers and the contribution of lamins dysfunction to disease are unarguable. However, the molecular mechanisms whereby lamins mutations cause pathologies remain less understood. Here, we review pathways and mechanisms recently identified as playing a role in the pathophysiology of laminopathies, with special emphasis in Hutchinson Gilford Progeria Syndrome (HGPS). This devastating incurable accelerated aging disease is caused by a silent mutation in the LMNA gene that generates a truncated lamin A protein "progerin" that exerts profound cellular toxicity and organismal decline. Patients usually die in their teens due to cardiovascular complications such as myocardial infarction or stroke. To date, there are no efficient therapies that ameliorate disease progression, stressing the need to understand molecularly disease mechanisms that can be targeted therapeutically. We will summarize data supporting that replication stress is a major cause of genomic instability in laminopathies, which contributes to the activation of innate immune responses to self-DNA that in turn accelerate the aging process.
Collapse
|
2048
|
Li L, Jiang Y, Lin H, Shen H, Sohn J, Alexander PG, Tuan RS. Muscle injury promotes heterotopic ossification by stimulating local bone morphogenetic protein-7 production. J Orthop Translat 2019; 18:142-153. [PMID: 31508317 PMCID: PMC6718974 DOI: 10.1016/j.jot.2019.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/03/2019] [Accepted: 06/05/2019] [Indexed: 02/01/2023] Open
Abstract
Background Heterotopic ossification (HO) is a pathological condition of abnormal bone formation in soft tissue, which causes pain and restricted range of motion in patients. There are two broad categories of HO, hereditary and acquired. Although different types of HO do not use identical mechanistic pathways of pathogenesis, muscle injury appears to be a unifying feature for all types of HO. However, little is known about the mechanisms by which muscle injury facilitates HO formation. Objective and method This study aimed to explore the cellular and molecular mechanisms linking muscle injury to HO by using cardiotoxin to induce muscle injury in a bone morphogenetic protein-2 (BMP-2)-induced HO mouse model. Results We found that muscle injury augmented HO formation and that this effect was correlated with BMP signalling activation and upregulation of BMP-7 expression at the early phase of HO progression. We further demonstrated that inhibition of BMP-7 activity in vitro suppressed the osteogenesis-promoting effect of conditioned medium derived from injured muscle tissue and in vivo reduced the volume of HO formation. We also showed that antiinflammatory drug treatment reduced the volume of HO with concomitant reduction in BMP-7 production. Conclusion In summary, our study has identified BMP-7 as a key osteoinductive factor in injured muscle that facilitates HO formation. The translational potential of this article Our results provide a candidate mechanistic rationale for the use of antiinflammatory drugs in the prevention of HO.
Collapse
Affiliation(s)
- La Li
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Graduate Program of Cellular and Molecular Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yangzi Jiang
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - He Shen
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jihee Sohn
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peter G. Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rocky S. Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Graduate Program of Cellular and Molecular Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Corresponding author. Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
2049
|
Baugh L, Watson MC, Kemmerling EC, Hinds PW, Huggins GS, Black LD. Knockdown of CD44 expression decreases valve interstitial cell calcification in vitro. Am J Physiol Heart Circ Physiol 2019; 317:H26-H36. [PMID: 30951363 PMCID: PMC6692733 DOI: 10.1152/ajpheart.00123.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/23/2022]
Abstract
The lack of pharmaceutical targets available to treat patients with calcific aortic valve disease (CAVD) necessitates further research into the specific mechanisms of the disease. The significant changes that occur to the aortic valves extracellular matrix (ECM) during the progression of CAVD suggests that these proteins may play an important role in calcification. Exploring the relationship between valve interstitial cells (VICs) and the ECM may lead to a better understand of CAVD mechanisms and potential pharmaceutical targets. In this study, we look at the effect of two ECM components, collagen and hyaluronic acid (HA), on the mineralization of VICs within the context of a two-dimensional, polyacrylamide (PAAM) model system. Using a novel, nondestructive imaging technique, we were able to track calcific nodule development in culture systems over a 3-wk time frame. We saw a significant increase in the size of the nodules grown on HA PAAM gels as compared with collagen PAAM gels, suggesting that HA has a direct effect on mineralization. Directly looking at the two known receptors of HA, CD44 and receptor for HA-mediated motility (RHAMM), and using siRNA knockdown revealed that a decrease in CD44 expression resulted in a reduction of calcification. A decrease in CD44, through siRNA knockdown, reduces mineralization on HA PAAM gels, suggesting a potential new target for CAVD treatment. NEW & NOTEWORTHY Our in vitro model of calcific aortic valve disease shows an interaction between the hyaluronic acid binding protein CD44 with the osteogenic factor OPN as a potential mechanism of aortic valve calcification. Using siRNA knockdown of CD44, we show an upregulation of OPN expression with a decrease in overall mineralization.
Collapse
Affiliation(s)
- Lauren Baugh
- Department of Biomedical Engineering, Tufts University , Medford, Massachusetts
| | - Matthew C Watson
- Department of Biomedical Engineering, Tufts University , Medford, Massachusetts
- Department of Mechanical Engineering, Tufts University , Medford, Massachusetts
| | - Erica C Kemmerling
- Department of Mechanical Engineering, Tufts University , Medford, Massachusetts
| | - Philip W Hinds
- Cellular, Molecular, and Developmental Biology Program, Sackler School for Graduate Biomedical Sciences, Tufts University School of Medicine , Boston, Massachusetts
| | - Gordon S Huggins
- Molecular Cardiology Research Center, Tufts Medical Center and Tufts University Sackler School for Graduate Biomedical Sciences , Boston, Massachusetts
| | - Lauren D Black
- Department of Biomedical Engineering, Tufts University , Medford, Massachusetts
- Cellular, Molecular, and Developmental Biology Program, Sackler School for Graduate Biomedical Sciences, Tufts University School of Medicine , Boston, Massachusetts
| |
Collapse
|
2050
|
Ren X, Yang C, Zhang L, Li S, Shi S, Wang R, Zhang X, Yue T, Sun J, Wang J. Copper metal-organic frameworks loaded on chitosan film for the efficient inhibition of bacteria and local infection therapy. NANOSCALE 2019; 11:11830-11838. [PMID: 31184673 DOI: 10.1039/c9nr03612a] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Although multiple advanced antibacterial and sterilization materials are available, bacterial infections still remain a big challenge in wound healing as they usually induce serious complications and cannot be thoroughly eliminated. Herein, we report an antibacterial film composed of the naturally derived polysaccharide chitosan (CS) and a copper metal-organic framework (HKUST-1) as a multifunctional platform for antibacterial and local infection therapy applications. As expected, the as-prepared HKUST-1/CS film possessed versatile abilities such as slow release of copper ions and reduced cytotoxicity; moreover, fluorescent staining and morphological changes of the bacteria treated with the HKUST-1/CS film confirmed the antibacterial activity of the fabricated film. Furthermore, in vivo results revealed that the HKUST-1/CS film could simultaneously kill bacteria and promote vessel regeneration; this resulted in an enhanced wound closure rate during the local infection therapy process. Overall, these results highlight that the HKUST-1/CS film exhibits significant potential as a suitable and promising wound dressing.
Collapse
Affiliation(s)
- Xinyi Ren
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, P. R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|