1
|
Patten J, Halligan P, Bashiri G, Kegel M, Bonadio JD, Wang K. EDA Fibronectin Microarchitecture and YAP Translocation during Wound Closure. ACS Biomater Sci Eng 2025; 11:2249-2262. [PMID: 40029610 DOI: 10.1021/acsbiomaterials.4c02019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Fibronectin (Fn) is an extracellular matrix glycoprotein with mechanosensitive structure-function. Extra domain A (EDA) Fn, a Fn isoform, is not present in adult tissue but is required for tissue repair. Curiously, EDA Fn is linked to both regenerative and fibrotic tissue repair. Given that Fn mechanoregulates cell behavior, EDA Fn organization during wound closure might play a role in mediating these differing responses. One mechanism by which cells sense and respond to their microenvironment is by activating a transcriptional coactivator, yes-associated protein (YAP). Interestingly, YAP activity is not only required for wound closure but similarly linked to both regenerative and fibrotic repair. Therefore, this study aims to evaluate how, during normal and fibrotic wound closure, EDA Fn organization might modulate YAP translocation by culturing human dermal fibroblasts on polydimethylsiloxane substrates mimicking normal (soft: 18 kPa) and fibrotic (stiff: 146 kPa) wounded skin. On stiffer substrates mimicking fibrotic wounds, fibroblasts assembled an aligned EDA Fn matrix comprising thinner fibers, suggesting increased microenvironmental tension. To evaluate if cell binding to the EDA domain of Fn was essential to overall matrix organization, fibroblasts were treated with Irigenin, which inhibits binding to the EDA domain within Fn. Blocking adhesion to EDA led to randomly organized EDA Fn matrices with thicker fibers, suggesting reduced microenvironmental tension even during fibrotic wound closure. To evaluate whether YAP signaling plays a role in EDA Fn organization, fibroblasts were treated with CA3, which suppresses YAP activity in a dose-dependent manner. Treatment with CA3 also led to randomly organized EDA Fn matrices with thicker fibers, suggesting a potential connected mechanism of reducing tension during fibrotic wound closure. Next, YAP activity was assessed to evaluate the impact of EDA Fn organization. Interestingly, fibroblasts migrating on softer substrates mimicking normal wounds increased YAP activity, but on stiffer substrates, they decreased YAP activity. When fibroblasts on stiffer substrates were treated with Irigenin or CA3, fibroblasts increased YAP activity. These results suggest that there may be disrupted signaling between EDA Fn organization and YAP translocation during fibrotic wound closure that could be restored when reestablishing normal EDA Fn matrix organization to instead drive regenerative wound repair.
Collapse
Affiliation(s)
- Jennifer Patten
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Patrick Halligan
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Michael Kegel
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Jacob D Bonadio
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
2
|
Yuan H, Wang X, Du S, Li M, Zhu E, Zhou J, Dong Y, Wang S, Shan L, Liu Q, Wang B. NELL2, a novel osteoinductive factor, regulates osteoblast differentiation and bone homeostasis through fibronectin 1/integrin-mediated FAK/AKT signaling. Bone Res 2025; 13:46. [PMID: 40210857 PMCID: PMC11986068 DOI: 10.1038/s41413-025-00420-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 04/12/2025] Open
Abstract
Neural EGFL-like 2 (NELL2) is a secreted protein known for its regulatory functions in the nervous and reproductive systems, yet its role in bone biology remains unexplored. In this study, we observed that NELL2 was diminished in the bone of aged and ovariectomized (OVX) mice, as well as in the serum of osteopenia and osteoporosis patients. In vitro loss-of-function and gain-of-function studies revealed that NELL2 facilitated osteoblast differentiation and impeded adipocyte differentiation from stromal progenitor cells. In vivo studies further demonstrated that the deletion of NELL2 in preosteoblasts resulted in decreased cancellous bone mass in mice. Mechanistically, NELL2 interacted with the FNI-type domain located at the C-terminus of Fibronectin 1 (Fn1). Moreover, we found that NELL2 activated the focal adhesion kinase (FAK)/AKT signaling pathway through Fn1/integrin β1 (ITGB1), leading to the promotion of osteogenesis and the inhibition of adipogenesis. Notably, administration of NELL2-AAV was found to ameliorate bone loss in OVX mice. These findings underscore the significant role of NELL2 in osteoblast differentiation and bone homeostasis, suggesting its potential as a therapeutic target for managing osteoporosis.
Collapse
Affiliation(s)
- Hairui Yuan
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
| | - Xinyu Wang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Shuanglin Du
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Mengyue Li
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Endong Zhu
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Jie Zhou
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Yuan Dong
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Shuang Wang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Liying Shan
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Qian Liu
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Baoli Wang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
| |
Collapse
|
3
|
Jin Z, Dai W, Huang Z, Li Q, Zhu Y, Wang W, Xu H. Engineered Titanium Oxide Nanoplatform for Targeted Photodynamic/Photothermal-Gas Therapy in Keloid Treatment. ACS APPLIED MATERIALS & INTERFACES 2025; 17:20705-20716. [PMID: 40138575 DOI: 10.1021/acsami.4c22289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Keloids pose a considerable worldwide health issue owing to their continual proliferation, invasiveness, and elevated recurrence rates. Keloids are abnormal scars formed through dysregulated wound healing processes, characterized by excessive keloid fibroblast (KF) proliferation, irregular collagen deposits, and persistent reticular dermis inflammation, which can lead to limited joint mobility, psychological distress, and severe pain and itching. In this study, we present metal-organic framework (MOF)-derived TiO2-based nanoparticles (LA@CTx NPs) synthesized as a phototherapy-gas-therapy nanoplatform, which have the ability to break down collagen, reduce inflammation, and stop the overproliferation of keloid fibroblasts. The MIL-125-derived nanoparticles maintain their crystalline framework while being rich in oxygen vacancies (OVs) and l-arginine (LA), enabling efficient photothermal conversion and reactive oxygen species (ROS) generation driven by synergistic near-infrared (NIR). Importantly, ROS generated by the NPs can trigger nitric oxide (NO) production by oxidizing LA, with the concentration of NO finely tunable via modulation of light conditions. This allows for a dual therapeutic effect: low NO concentrations suppress inflammation, while higher concentrations induce cell death. In vitro and in vivo investigations show that LA@CTx nanoparticles efficiently eliminate primary keloid lesions and provoke apoptosis in keloid cells by dual-modality activation of photodynamic and photothermal treatments facilitated by single NIR irradiation. The study presents an innovative method of therapy for the clinical treatment of keloids.
Collapse
Affiliation(s)
- Zilong Jin
- The Education Ministry Key Laboratory of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry of Ministry of Education, Shanghai Key Laboratory of Rare Earth Functional Materials, and Shanghai Frontiers Science Center of Biomimetic Catalysis, Shanghai Normal University, Shanghai 200234, People's Republic of China
| | - Wufei Dai
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Department of Medicine 1, HoUniversity spital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen 91054, Germany
| | - Zhengjie Huang
- The Education Ministry Key Laboratory of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry of Ministry of Education, Shanghai Key Laboratory of Rare Earth Functional Materials, and Shanghai Frontiers Science Center of Biomimetic Catalysis, Shanghai Normal University, Shanghai 200234, People's Republic of China
| | - Qinglin Li
- Research Institute of Plastic Surgery, Shandong Second Medical University, Weifang, Shandong 261000, China
| | - Yuduo Zhu
- The Education Ministry Key Laboratory of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry of Ministry of Education, Shanghai Key Laboratory of Rare Earth Functional Materials, and Shanghai Frontiers Science Center of Biomimetic Catalysis, Shanghai Normal University, Shanghai 200234, People's Republic of China
| | - Wenbo Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - He Xu
- The Education Ministry Key Laboratory of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry of Ministry of Education, Shanghai Key Laboratory of Rare Earth Functional Materials, and Shanghai Frontiers Science Center of Biomimetic Catalysis, Shanghai Normal University, Shanghai 200234, People's Republic of China
| |
Collapse
|
4
|
Chocarro-Wrona C, Pleguezuelos-Beltrán P, López de Andrés J, Antich C, de Vicente J, Jiménez G, Arias-Santiago S, Gálvez-Martín P, López-Ruiz E, Marchal JA. A bioactive three-layered skin substitute based on ECM components effectively promotes skin wound healing and regeneration. Mater Today Bio 2025; 31:101592. [PMID: 40092225 PMCID: PMC11910132 DOI: 10.1016/j.mtbio.2025.101592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
To overcome the limitations of conventional skin tissue engineering (TE), 3D biofabrication approaches are being developed. However, tissue mimicry should be further improved in skin models. Here, we developed and characterized biomimetic hydrogels to obtain a biofabricated three-layered (BT) skin substitute based on the main components found in the epidermal, dermal, and hypodermal skin layers. Hydrogels for dermal and hypodermal skin layers were based on a mix of agarose and type I collagen, supplemented with skin-related extracellular matrix (ECM) components (dermatan sulfate, hyaluronic acid, and elastin) and loaded with human dermal fibroblasts (hDFs) or human mesenchymal stem/stromal cells (hMSCs), respectively. The epidermal hydrogel was formulated using type I collagen supplemented with keratin and sphingolipids, and seeded with human epidermal keratinocytes (hEKs). Physicochemical results revealed adequate viscosity, gelling times, and pH for each hydrogel solution. The BT Skin also showed good swelling and degradation kinetics, and mechanical properties in a similar range of human skin. The hydrogels and BT Skin demonstrated stable cell viability and metabolic activity, as well as intercellular communication through the release of growth factors. Moreover, the BT Skin demonstrated controlled inflammation in vivo, and produced results comparable to autografting in a mouse skin wound model. This bioactive and biomimetic three-layered BT Skin has a composition that attempts to mimic the natural ECM of the skin, formulated with the characteristic cells and biomolecules present in each skin layer, and offers promising properties for its clinical application in the treatment of patients with skin injuries.
Collapse
Affiliation(s)
- Carlos Chocarro-Wrona
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016, Granada, Spain
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18016, Granada, Spain
| | - Paula Pleguezuelos-Beltrán
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016, Granada, Spain
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18016, Granada, Spain
| | - Julia López de Andrés
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016, Granada, Spain
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18016, Granada, Spain
| | - Cristina Antich
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18016, Granada, Spain
- National Center for Advancing Translational Sciences, National Institute of Health, 28050, Rockville, MD, USA
| | - Juan de Vicente
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- F2N2Lab, Magnetic Soft Matter Group, Department of Applied Physics, Faculty of Sciences, University of Granada, 18071, Granada, Spain
| | - Gema Jiménez
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016, Granada, Spain
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18016, Granada, Spain
| | - Salvador Arias-Santiago
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Dermatology Department, Hospital Universitario Virgen de las Nieves, 18012, Granada, Spain
- Dermatology Department, Faculty of Medicine, University of Granada, 18016, Granada, Spain
| | | | - Elena López-Ruiz
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18016, Granada, Spain
- Department of Health Sciences, University of Jaén, 23071, Jaén, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Center for Biomedical Research (CIBM), University of Granada, 18016, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, 18012, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016, Granada, Spain
- Excellence Research Unit “Modelling Nature” (MNat), University of Granada, 18016, Granada, Spain
- BioFab i3D - Biofabrication and 3D (bio)printing laboratory, University of Granada, 18016, Granada, Spain
| |
Collapse
|
5
|
Wang J, Xu M, Liu H, Wang D, Zhang H, Xu Z, Shi X, Liu X, Tan Z. Vascular grafts with a mimetic microenvironment extracted from extracellular matrix of adipocytes can promote endothelialization in vivo. Acta Biomater 2025:S1742-7061(25)00222-3. [PMID: 40169081 DOI: 10.1016/j.actbio.2025.03.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/03/2025]
Abstract
Synthetic vascular substitutes are widely studied for small-caliber arteries replacement but their efficacy requires further improvement. Vascular tissue engineering holds great promise for preparing small-caliber vascular grafts with therapeutic effects, and previous work has demonstrated that the cellular layer at the luminal surface of vascular grafts has the potential to provide high functionality to vascular tissue. Improved endothelialization has been proven to be a key strategy for promoting the efficacy of vascular regeneration. However, there still remains a challenge of finding proper endothelialization methods or cell types to guarantee vascular grafts the long-term patency and functions. Herein, a biomimetic bilayer vascular graft was developed by 3D printing and electrospinning techniques. The electrospun PCL nanofiber was fabricated as the outer supporting layer while a biomimetic inner layer structure composed of cell extracellular matrix microenvironment was prepared by a decellularization process. This inner layer was designed to favor endothelial cell (EC) adhesion and enhance endothelialization on the surfaces of vascular grafts. Fibronectin, derived from adipocytes, provided a naturally occurring substrate for EC adhesion. The findings showed that by binding fibronectin, integrin α5β1 mediates EC adherence to the designed vascular graft. The bilayer graft with a mimetic microenvironment extracted from extracellular matrix of adipocytes can promote endothelialization and sustain good patency in vivo, which may represent a promising biomaterial for clinical vascular transplantation. STATEMENT OF SIGNIFICANCE: This study proposed a universal method for including any substrate type in vascular cell type-specific extracellular matrices (ECM) via regulating selective adhesion to promote vascular tissue regeneration. The reconstructed 3D ECM recapitulating a vascular-like microenvironment promoted the orderly regeneration and functional recovery of vascular tissues in vivo. The findings represent a proof of principle for vascular cell selectivity in cell type-specific ECM microenvironments, and provide a valuable perspective for further investigations on the controlled regeneration of heterogeneous tissues.
Collapse
Affiliation(s)
- Jian Wang
- College of Biology, Hunan University, Changsha 410082, China; Institute of Shenzhen, Hunan University, Shenzhen 518000, China
| | - Miaomiao Xu
- College of Biology, Hunan University, Changsha 410082, China
| | - Hui Liu
- College of Biology, Hunan University, Changsha 410082, China; Institute of Shenzhen, Hunan University, Shenzhen 518000, China
| | - Danling Wang
- College of Biology, Hunan University, Changsha 410082, China; Greater Bay Area Institute for Innovation, Hunan University, Guangzhou 511300, China
| | - Hengyuan Zhang
- College of Biology, Hunan University, Changsha 410082, China; Greater Bay Area Institute for Innovation, Hunan University, Guangzhou 511300, China
| | - Zilong Xu
- College of Biology, Hunan University, Changsha 410082, China; Greater Bay Area Institute for Innovation, Hunan University, Guangzhou 511300, China
| | - Xiuyuan Shi
- College of Biology, Hunan University, Changsha 410082, China; Greater Bay Area Institute for Innovation, Hunan University, Guangzhou 511300, China
| | - Xiao Liu
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou 511300, China.
| | - Zhikai Tan
- College of Biology, Hunan University, Changsha 410082, China; Institute of Shenzhen, Hunan University, Shenzhen 518000, China; Greater Bay Area Institute for Innovation, Hunan University, Guangzhou 511300, China.
| |
Collapse
|
6
|
Ouyang P, Qi J, Tong B, Li Y, Cao J, Wang L, Niu T, Qi X. Butyrate Ameliorates Graves' Orbitopathy Through Regulating Orbital Fibroblast Phenotypes and Gut Microbiota. Invest Ophthalmol Vis Sci 2025; 66:5. [PMID: 40035727 DOI: 10.1167/iovs.66.3.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025] Open
Abstract
Purpose Graves' orbitopathy (GO), the common extrathyroidal complication of Graves' disease (GD), is characterized by orbital fibroblast stimulation, adipogenesis, and hyaluronan production. Recently, gut microbiota and its metabolites have garnered attention for their possible involvement in GO. Methods This study utilized an animal model of GO and examined the effects of butyrate treatment on orbital fibroblast cells and gut microbiota. Ex vivo experiments were performed using orbital fibroblasts derived from healthy patients' and patients' with GO orbital tissue to evaluate vitality, activation, and adipogenesis in response to butyrate treatment. Gut microbiota diversity was also analyzed in butyrate-treated and untreated GO mice. Results In human orbital fibroblasts, butyrate treatment dramatically decreased the vitality of GO-derived fibroblasts without harming normal fibroblasts. Butyrate prevented activation and fibrotic processes induced by transforming growth factor beta 1 (TGF-β1) in GO and normal fibroblasts. Additionally, butyrate reduced lipid droplet formation and downregulated lipogenic markers in GO and normal orbital fibroblasts, inhibiting adipogenesis. In the GO mouse model, butyrate therapy improved orbital histological abnormalities and normalized serum thyroid hormone and antibody levels. The intestinal microbiome of butyrate-treated GO mice also changed significantly, with a reduction in certain bacteria (Bifidobacterium, GCA-900066575, and Parabacteroides) and an increase in others (Bacteroides and Rikenellaceae_RC9). Conclusions Butyrate ameliorates several of the symptoms of GO, lowering GO orbital fibroblast viability, adipogenesis, and TGF-β1-induced fibrosis without damaging normal fibroblasts. Butyrate normalizes thyroid function in a GO mouse model, improves histopathological alterations, and transforms gut microbiota populations, proving its potential in treating GO through the gut-thyroid axis.
Collapse
Affiliation(s)
- Pingbo Ouyang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jia Qi
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Boding Tong
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yunping Li
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiamin Cao
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lujue Wang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tongxin Niu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Qi
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
7
|
Al-Shamaa RM, Al-Askary RA. Remineralizing capacity of zinc oxide eugenol sealer following the addition of nanohydroxyapatite-tyrosine amino acid: An in vivo animal study. J Oral Biosci 2025; 67:100567. [PMID: 39419338 DOI: 10.1016/j.job.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE Although several sealers have been developed, the zinc oxide eugenol (ZOE) sealer is still used in many private dental clinics. This study aimed to compare the biocompatibility and remineralizing capacity οf ZOE sealer with the addition of nanohydroxyapatite-tyrosine amino acid. METHODS This study was conducted on Twenty rabbits. The rabbits were divided into four groups based on the test observation period (3, 7, 21, and 28 days) following surgical implantation. General anesthesia was administered to each rabbit, and a subcutaneous incision of approximately 1 ± 0.5 cm was made along the symphyseal area of the mandible of each rabbit. Four bone cavities were generated in the interdental space of the lower jaw between the central and molar teeth of each rabbit, with one longitudinal subcutaneous incision. The ZOE sealers were mixed according to the manufacturer's guidelines and directly inserted within the cavities generated at the end of each test period. The animals were sacrificed, and bone biopsy was carried out at the site of testing. The biopsy samples were obtained and subjected to histological analysis using a low-power light microscope (Olympus C ⅹ 21, Japan) and immunohistochemistry using Ki67 antibody. RESULTS The collected information was examined by parametric statistical tests using SPSS software version ''22." One-way ANOVA and post-hoc Duncan's tests were used to measure the significance among various groups, with statistical significance set, when "P ≤ 0.01". CONCLUSION The 20% mixed endodontic sealer displayed excellent outcomes compared to other experimental groups, as identified by higher new bone formation at every evaluation period.
Collapse
Affiliation(s)
- Rasha M Al-Shamaa
- Department of Conservative Dentistry, College of Dentistry, Mosul University, 41001, Iraq.
| | - Raghad A Al-Askary
- Department of Conservative Dentistry, College of Dentistry, Mosul University, 41001, Iraq.
| |
Collapse
|
8
|
Zhao P, Li Y, Guo B, Liu Z, Zhang X, Liu M, Ma X. Hydrogen-Releasing Micromaterial Dressings: Promoting Wound Healing by Modulating Extracellular Matrix Accumulation Through Wnt/β-Catenin and TGF-β/Smad Pathways. Pharmaceutics 2025; 17:279. [PMID: 40142944 PMCID: PMC11944919 DOI: 10.3390/pharmaceutics17030279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Wound healing is a complex and intricate biological process that involves multiple systems within the body and initiates a series of highly coordinated responses to repair damage and restore integrity and functionality. We previously identified that breathing hydrogen can significantly inhibit early inflammation, activate autologous stem cells, and promote the accumulation of extracellular matrix (ECM). However, the broader functions and downstream targets of hydrogen-induced ECM accumulation and tissue remodeling are unknown in the wound-healing process. Methods: Consequently, this thesis developed a hydrogen sustained-release dressing based on a micro storage material and reveals the mechanism of hydrogen in treating wound healing. Upon encapsulating the hydrogen storage materials, magnesium (Mg), and ammonia borane (AB), we found that SiO2@Mg exhibits superior sustained-release performance, while SiO2@AB demonstrates a higher hydrogen storage capacity. We used a C57/BL6 mouse full-thickness skin defect wound model to analyze and compare different hydrogen dressings. Results: It was identified that hydrogen dressings can significantly improve the healing rate of wounds by promoting epithelialization, angiogenesis, and collagen accumulation in wound tissue, and that the effect of slow-release dressings is better than of non-slow-release dressings. We also found that hydrogen dressing can promote transcriptome-level expression related to cell proliferation and differentiation and ECM accumulation, mainly through the Wnt1/β-catenin pathway and TGF-β1/Smad2 pathway. Conclusions: Overall, these results provide a novel insight into the field of hydrogen treatment and wound healing.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xuemei Ma
- College of Chemistry and Life Sciences, Beijing University of Technology, Beijing 100124, China; (P.Z.); (Y.L.); (B.G.); (Z.L.); (X.Z.); (M.L.)
| |
Collapse
|
9
|
Hartley A, Williams PM, Mata A. A Comparison of the Mechanical Properties of ECM Components and Synthetic Self-Assembling Peptides. Adv Healthc Mater 2025:e2402385. [PMID: 39972630 DOI: 10.1002/adhm.202402385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 01/20/2025] [Indexed: 02/21/2025]
Abstract
The field of tissue engineering is increasingly moving away from a one-size-fits-all approach of simple synthetic homogeneous gels, and embracing more tailored designs to optimize cell function and differentiation for the organ of interest. Extracellular matrix (ECM) proteins are still the optimal route for controlling cell function, while a field of great promise is that of synthetic self-assembling peptides (SSAPs), which are fully biocompatible, biodegradable, and offer both the hierarchical structure and dynamic properties displayed by protein networks found in natural tissue. However, the mechanical properties of neither group have been comprehensively reviewed. In this review, rheological data and the Young's modulus of the most prevalent proteins involved in the ECM (collagen I, elastin, and fibronectin) are collated for the first time, and compared against the most widely researched SSAPs: peptide amphiphiles (PAs), β-sheets, β-hairpin peptides, and Fmoc-based gels (with a focus on PA-E3, RADA16, MAX1, and FmocFF, respectively).
Collapse
Affiliation(s)
- Alex Hartley
- School of Pharmacy, University of Nottingham, University Park Campus, Nottingham, NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, University Park Campus, Nottingham, NG7 2RD, UK
| | - Philip Michael Williams
- School of Pharmacy, University of Nottingham, University Park Campus, Nottingham, NG7 2RD, UK
| | - Alvaro Mata
- School of Pharmacy, University of Nottingham, University Park Campus, Nottingham, NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, University Park Campus, Nottingham, NG7 2RD, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, University Park Campus, Nottingham, NG7 2RD, UK
| |
Collapse
|
10
|
Rossi F, Luppi S, Fejza A, Giolo E, Ricci G, Andreuzzi E. Extracellular matrix and pregnancy: functions and opportunities caught in the net. Reprod Biol Endocrinol 2025; 23:24. [PMID: 39953593 PMCID: PMC11827249 DOI: 10.1186/s12958-025-01348-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/24/2025] [Indexed: 02/17/2025] Open
Abstract
The extracellular matrix is a complex network of macromolecules that support the growth and homeostatic development of organisms. By conveying multiple signaling cascades, it impacts on several biological processes and influences the behaviour of numerous cell types. During the endometrial cycle and the key events necessary for a correct embryo implantation and placentation, this bioactive meshwork is substantially modified to favour endometrial receptivity and vascular adaptation, trophoblast cell migration, and immune activation as well. A correct extracellular remodeling is fundamental for the establishment of a physiological pregnancy; indeed, the occurrence of altered matrix modifications associates with gestational disorders such as preeclampsia. In the present review, we will critically evaluate the role of pivotal matrix constituents in regulating the key steps of embryo implantation and placentation, provide up-to-date information concerning their primary mechanisms of action and discuss on their potential as a novel source of biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Francesca Rossi
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, 34137, Italy
| | - Stefania Luppi
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, 34137, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, 34127, Italy
| | - Albina Fejza
- UBT-Higher Education Institution, Kalabria, Street Rexhep Krasniqi Nr. 56, Prishtina, 10000, Kosovo
| | - Elena Giolo
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, 34137, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, 34137, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, 34127, Italy
| | - Eva Andreuzzi
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, 34137, Italy.
| |
Collapse
|
11
|
Rappold R, Kalogeropoulos K, La Regina G, auf dem Keller U, Slack E, Vogel V. Relaxation of mucosal fibronectin fibers in late gut inflammation following neutrophil infiltration in mice. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:4. [PMID: 39917413 PMCID: PMC11794144 DOI: 10.1038/s44341-024-00006-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/13/2024] [Indexed: 02/09/2025]
Abstract
The continuously remodeled extracellular matrix (ECM) plays a pivotal role in gastrointestinal health and disease, yet its precise functions remain elusive. In this study, we employed laser capture microdissection combined with low-input proteomics to investigate ECM remodeling during Salmonella-driven inflammation. To complement this, we probed how fibronectin fiber tension is altered using a mechanosensitive peptide probe. While fibronectin fibers in healthy intestinal tissue are typically stretched, many lose their tension in intestinal smooth muscles only hours after infection, despite the absence of bacteria in that area. In contrast, within the mucosa, where Salmonella is present starting 12 h post infection, fibronectin fiber relaxation occurred exclusively during late-stage infection at 72 h and was localized to already existing clusters of infiltrated neutrophils. Using N-terminomics, we identified three new cleavage sites in fibronectin in the inflamed cecum. The unique, tissue layer-specific changes in the molecular compositions and ECM fiber tension revealed herein might trigger new therapeutic strategies to fight acute intestinal inflammation.
Collapse
Affiliation(s)
- Ronja Rappold
- Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | | | - Gianna La Regina
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Ulrich auf dem Keller
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Emma Slack
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
- Botnar Research Center for Child Health, Basel, Switzerland
| | - Viola Vogel
- Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
- Botnar Research Center for Child Health, Basel, Switzerland
| |
Collapse
|
12
|
Wang Y, Hess ME, Tan Y, Esser PR, Nyström A, Boerries M, Sayar SB, Has C. Alterations in the microenvironment of junctional epidermolysis bullosa keratinocytes: A gene expression study. Matrix Biol 2025; 135:12-23. [PMID: 39615637 DOI: 10.1016/j.matbio.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/12/2024] [Accepted: 11/24/2024] [Indexed: 01/15/2025]
Abstract
Integrin α6β4 subunits and type XVII collagen are critical transmembrane proteins involved in cell-matrix adhesion in skin, while laminin 332 serves as their ligand in the basement membrane zone (BMZ). Those proteins contribute to the composition of hemidesmosomes (HDs) and pathogenic variants in their corresponding genes cause junctional epidermolysis bullosa (JEB). Although the genotype-phenotype relationships in JEB have been extensively studied, the pathogenetic changes of extracellular matrix (ECM) and cell-matrix adhesion resulting from gene mutations remain unclear. We conducted a global unbiased transcriptome analysis using bulk RNA sequencing (RNA-seq) on selected JEB donor-derived cell lines lacking integrin β4 subunit (ITGB4-), type XVII collagen (COL17-) and laminin β3 chain (LAMB3-), respectively. Additional JEB cell lines and JEB donor skin samples were used for validation of relevant findings. Collectively, the results revealed similar dysregulation patterns of ECM and focal adhesion (FAs) associated genes in ITGB4- and COL17- cell lines, while LAMB3- cells displayed a relatively opposite tendency. Importantly, key nodes in the dysregulated network were associated with ECM proteins involved in wound healing processes. Additionally, a group of inflammatory-associated genes was disclosed to be up-regulated in JEB keratinocytes and could not be normalized by the adhesion rescue. The functional assay further revealed the hierarchy of stable adhesion among mutant cell lines COL17->ITGB4->LAMB3-, which correlates with the severity of their clinical manifestations. Our results indicated a wound healing associated ECM and inflammatory microenvironment established by JEB keratinocytes.
Collapse
Affiliation(s)
- Yao Wang
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Maria Elena Hess
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Yan Tan
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Philipp R Esser
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Alexander Nyström
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Comprehensive Cancer Centre Freiburg (CCCF), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; German Cancer Consortium (DKTK), a partnership between DKFZ and Medical Center - University of Freiburg, Freiburg, Germany
| | - Saliha Beyza Sayar
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Cristina Has
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
13
|
Ye S, Chen B, Jeevithan L, Yang H, Kong Y, Diao X, Wu W. Recombinant Humanized Collagen Enhances Secreted Protein Levels of Fibroblasts and Facilitates Rats' Skin Basement Membrane Reinforcement. J Funct Biomater 2025; 16:47. [PMID: 39997581 PMCID: PMC11856143 DOI: 10.3390/jfb16020047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/25/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Collagen and its peptides exhibit remarkable antioxidant activity, superior biocompatibility, and water solubility, making them a significant research focus in skin care. Hence, the recombinant humanized collagen types I, III, and XVII complexed with niacinamide were developed to address damage in human foreskin fibroblasts (HFF-1) caused by ultraviolet radiation and to evaluate basement membrane proteins in a rat skin model. The Cell Counting Kit-8 (CCK-8) assay showed that higher concentrations of the complex increased the survival of damaged cells by approximately 10% and 22%, respectively, compared to the normal group after 16 and 48 h of treatment. Further biochemical analyses using ELISA and immunofluorescence (IF) confirmed that the complex enhanced the expression of collagen type IV, laminin, P63, and transforming growth factor-β (TGF-β) in the damaged cells. Additionally, the complex boosted the activity of the basement membrane in rat skin and stimulated the secretion of integrin, laminin, and perlecan. Overall, the recombinant humanized collagen complex effectively reinforced the skin's basement membrane.
Collapse
Affiliation(s)
- Shijia Ye
- Department of Marine Biopharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (S.Y.); (B.C.); (L.J.); (H.Y.); (Y.K.)
| | - Boyu Chen
- Department of Marine Biopharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (S.Y.); (B.C.); (L.J.); (H.Y.); (Y.K.)
| | - Lakshmi Jeevithan
- Department of Marine Biopharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (S.Y.); (B.C.); (L.J.); (H.Y.); (Y.K.)
- Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM-Universidad Catolica San-Antonio de Murcia, 30107 Murcia, Spain
| | - Haoze Yang
- Department of Marine Biopharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (S.Y.); (B.C.); (L.J.); (H.Y.); (Y.K.)
| | - Yaqi Kong
- Department of Marine Biopharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (S.Y.); (B.C.); (L.J.); (H.Y.); (Y.K.)
| | - Xiaozhen Diao
- Department of Marine Biopharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (S.Y.); (B.C.); (L.J.); (H.Y.); (Y.K.)
- Putuo Branch of International Combined Research Center for Marine Biological Sciences, Zhoushan 316104, China
| | - Wenhui Wu
- Department of Marine Biopharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; (S.Y.); (B.C.); (L.J.); (H.Y.); (Y.K.)
- Putuo Branch of International Combined Research Center for Marine Biological Sciences, Zhoushan 316104, China
- Marine Biomedical Science and Technology Innovation Platform of Lin-Gang Special Area, Shanghai 201306, China
| |
Collapse
|
14
|
Kalmár T, Jakab D, Maróti Z, Pásztor G, Turkevi-Nagy S, Kemény É, Hopfer H, Becker JU, Bereczki C, Iványi B. Fibronectin Glomerulopathy Without Typical Renal Biopsy Features in a 4-Year-Old Girl with Incidentally Discovered Proteinuria and a G417V FN1 Gene Mutation. Int J Mol Sci 2025; 26:641. [PMID: 39859354 PMCID: PMC11766185 DOI: 10.3390/ijms26020641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Fibronectin glomerulopathy (FG) is caused by fibronectin 1 (FN1) gene mutations. A renal biopsy was performed on a 4-year-old girl with incidentally discovered proteinuria (150 mg/dL); her family history of renal disease was negative. Markedly enlarged glomeruli (mean glomerular diameter: 196 μm; age-matched controls: 140 μm), α-SMA-positive and Ki-67-positive mesangial cell proliferation (glomerular proliferation index 1.76), the mild expansion of mesangial areas, no immune or electron-dense deposits, normal glomerular basement membrane, and diffusely effaced foot processes were observed. Genetic testing identified a de novo heterozygous mutation (Gly417Val) in the collagen-binding site of the FN II-2 domain, prompting fibronectin immunostaining. Strong mesangial positivity was noted, hence FG was diagnosed. The follow-up period of 29 months revealed nephrotic range proteinuria, intermittent microhematuria, glomerular hyperfiltration, and preserved renal function. The biopsy features of early childhood-onset FG were compared to a case of FG with a lobular pattern diagnosed in a 44-year-old patient with undulating proteinuria, microhematuria, hypertension known for a year, and a positive family history. Early childhood-onset FG was characterized by glomerular enlargement, mesangial proliferation, and no changes that suggested fibronectin deposition disease. In summary, the novel aspects of the case were that the mutation was located at the collagen-binding site of the FN1 gene, not identified earlier, and the histologic spectrum of FG was expanded by the observed mesangial proliferative pattern and striking glomerulomegaly. Now, FG should also be considered among the monogenic causes of proteinuric kidney diseases in pediatric nephrology practice.
Collapse
Affiliation(s)
- Tibor Kalmár
- Genetic Diagnostic Laboratory, Department of Pediatrics, Albert Szent-Györgyi Medical Center, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Dániel Jakab
- Department of Pediatrics, Albert Szent-Györgyi Medical Center, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Zoltán Maróti
- Genetic Diagnostic Laboratory, Department of Pediatrics, Albert Szent-Györgyi Medical Center, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Gyula Pásztor
- Department of Radiology, Albert Szent-Györgyi Medical Center, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Sándor Turkevi-Nagy
- Department of Pathology, Albert Szent-Györgyi Medical Center, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary; (S.T.-N.); (B.I.)
| | - Éva Kemény
- Department of Pathology, Albert Szent-Györgyi Medical Center, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary; (S.T.-N.); (B.I.)
| | - Helmut Hopfer
- Institut für Pathologie, Unversitätsspital Basel, 4031 Basel, Switzerland
| | - Jan Ulrich Becker
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Uniklinik Köln, 50937 Köln, Germany
| | - Csaba Bereczki
- Department of Pediatrics, Albert Szent-Györgyi Medical Center, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Béla Iványi
- Department of Pathology, Albert Szent-Györgyi Medical Center, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary; (S.T.-N.); (B.I.)
| |
Collapse
|
15
|
Gillesberg FS, Pehrsson M, Bay-Jensen AC, Frederiksen P, Karsdal M, Deleuran BW, Kragstrup TW, Kubo S, Tanaka Y, Mortensen JH. Regulation of fibronectin and collagens type I, III and VI by TNF-α, TGF-β, IL-13, and tofacitinib. Sci Rep 2025; 15:1087. [PMID: 39774197 PMCID: PMC11707072 DOI: 10.1038/s41598-024-84151-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Understanding how inflammatory cytokines influence profibrogenic wound healing responses in fibroblasts is important for understanding the pathogenesis of fibrosis. TNF-α and IL-13 are key cytokines in Th1 and Th2 immune responses, respectively, while TGF-β1 is the principal pro-fibrotic mediator. We show that 12-day fibroblast culture with TNF-α or IL-13 induces fibrogenesis, marked by progressively increasing type III and VI collagen formation, and that TGF-β1 co-stimulation amplifies these effects. Tofacitinib substantially reduced the formation of ECM proteins in response to IL-13, while fibrogenesis in response to TNF-α or TGF-β1 was marginally inhibited. The in vitro findings were supported by clinical observations in patients with active rheumatoid arthritis, which had elevated serum type III collagen formation, indicating ongoing fibrogenesis during inflammation. After 48-60 weeks of tofacitinib treatment, type III collagen degradation, aswell as formation, were significantly decreased compared to baseline, highlighting dual anti-inflammatory and anti-fibrogenic effects of tofacitinib. In contrast, other anti-inflammatory treatments including methotrexate, adalimumab and tocilizumab demonstrated anti-inflammatory effects only. Our results highlight fibro-inflammatory profiles associated with TNF-α or IL-13 stimulation, both alone and in combination with TGF-β1, and support the use of tofacitinib as an anti-fibrogenic treatment in chronic inflammatory conditions.
Collapse
Affiliation(s)
- Frederik S Gillesberg
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark.
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, Aarhus C, 8000, Denmark.
| | - Martin Pehrsson
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark
| | | | - Peder Frederiksen
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark
| | - Morten Karsdal
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark
| | - Bent W Deleuran
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, Aarhus C, 8000, Denmark
- Department of Rheumatology, Århus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
| | - Tue W Kragstrup
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, Aarhus C, 8000, Denmark
- Department of Rheumatology, Århus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
- Diagnostic Center, Regional Hospital Silkeborg, Falkevej 1, Silkeborg, 8600, Denmark
| | - Satoshi Kubo
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi, 807-8555, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi, 807-8555, Kitakyushu, Japan
| | - Joachim H Mortensen
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark
| |
Collapse
|
16
|
Kim SE, Noda R, Liu YC, Nakajima Y, Kameoka S, Motooka D, Mizuno S, Takahashi S, Takaya K, Murase T, Ikematsu K, Tratsiakova K, Motoyama T, Nakashima M, Kishi K, Martin P, Seno S, Okuzaki D, Mori R. Novel integrated multiomics analysis reveals a key role for integrin beta-like 1 in wound scarring. EMBO Rep 2025; 26:122-152. [PMID: 39558136 PMCID: PMC11724056 DOI: 10.1038/s44319-024-00322-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 09/30/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024] Open
Abstract
Exacerbation of scarring can originate from a minority fibroblast population that has undergone inflammatory-mediated genetic changes within the wound microenvironment. The fundamental relationship between molecular and spatial organization of the repair process at the single-cell level remains unclear. We have developed a novel, high-resolution spatial multiomics method that integrates spatial transcriptomics with scRNA-Seq; we identified new characteristic features of cell-cell communication and signaling during the repair process. Data from PU.1-/- mice, which lack an inflammatory response, combined with scRNA-Seq and Visium transcriptomics, led to the identification of nine genes potentially involved in inflammation-related scarring, including integrin beta-like 1 (Itgbl1). Transgenic mouse experiments confirmed that Itgbl1-expressing fibroblasts are required for granulation tissue formation and drive fibrogenesis during skin repair. Additionally, we detected a minority population of Acta2high-expressing myofibroblasts with apparent involvement in scarring, in conjunction with Itgbl1 expression. IL1β signaling inhibited Itgbl1 expression in TGFβ1-treated primary fibroblasts from humans and mice. Our novel methodology reveal molecular mechanisms underlying fibroblast-inflammatory cell interactions that initiate wound scarring.
Collapse
Affiliation(s)
- Sang-Eun Kim
- Department of Pathology, School of Medicine, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Ryota Noda
- Department of Pathology, School of Medicine, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Yu-Chen Liu
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yukari Nakajima
- Department of Plastic and Reconstructive Surgery, School of Medicine, Keio University, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shoichiro Kameoka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Daisuke Motooka
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center, Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kento Takaya
- Department of Plastic and Reconstructive Surgery, School of Medicine, Keio University, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takehiko Murase
- Department of Forensic Pathology and Science, School of Medicine, Nagasaki University, Nagasaki, 852-8523, Japan
- Department of Forensic Medicine, Faculty of Medicine, Kagawa University, Kita, Kagawa, 761-0793, Japan
| | - Kazuya Ikematsu
- Department of Forensic Pathology and Science, School of Medicine, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Katsiaryna Tratsiakova
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Takahiro Motoyama
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Masahiro Nakashima
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Kazuo Kishi
- Department of Plastic and Reconstructive Surgery, School of Medicine, Keio University, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Paul Martin
- Department of Biochemistry, Biomedical Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Suita, Osaka, 565-0871, Japan.
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Suita, Osaka, 565-0871, Japan.
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan.
| | - Ryoichi Mori
- Department of Pathology, School of Medicine, Nagasaki University, Nagasaki, 852-8523, Japan.
- Department of Tissue Repair and Regenerative Medical Science, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, 852-8523, Japan.
- Leading Medical Research Core Unit, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8523, Japan.
| |
Collapse
|
17
|
Villata S, Baruffaldi D, Cue Lopez R, Paoletti C, Bosch P, Napione L, Giovannozzi AM, Pirri CF, Martinez-Campos E, Frascella F. Broadly Accessible 3D In Vitro Skin Model as a Comprehensive Platform for Antibacterial Therapy Screening. ACS APPLIED MATERIALS & INTERFACES 2024; 16:70284-70296. [PMID: 39667725 DOI: 10.1021/acsami.4c16397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Skin infections are currently a worldwide emergency as antibiotic-resistant bacteria are spreading, leading to the ineffectiveness of most antibiotics and antibacterial strategies. Consequently, there is an urgency of developing and testing innovative antibacterial therapies. As traditional 2D cell culture and planktonic bacteria culture can be obsolete due to their incapability of resembling the complex infection environment, 3D in vitro skin models can be a powerful tool to test and validate therapies. In this article, a 3D in vitro epidermis-dermis skin model has been developed and biofabricated to be broadly available, reaching a balance between the simplicity and reproducibility of the model and its complexity in terms of wound, infection, and treatment response. The results are really promising, as the skin model developed a comprehensive physical barrier. To further investigate the skin model, controlled wounding, infection, and antibiotic treatments were performed. The results were remarkable: Not only was the unwounded epidermal barrier able to partially stop the bacterial proliferation, but the entire system reacted to both wound and infection in a complex and complete way. Extracellular matrix deposition and remodeling, inflammatory response, antimicrobial peptide production, and change in cellular behaviors, from epithelial to mesenchymal and from fibroblasts to myofibroblasts, were witnessed, with different extents depending on the bacterial strain. In addition, the inflammatory response to the antibiotic administration was opposite for the two bacterial infections, probably revealing the release of inflammatory endotoxins during Escherichia coli death. In conclusion, the presented 3D in vitro skin model has all the characteristics to be a future landmark as a platform for antibacterial strategy therapy testing.
Collapse
Affiliation(s)
- Simona Villata
- Dipartimento di Scienza Applicata e Tecnologia, PolitoBIOMed Lab, Politecnico di Torino, Turin 10129, Italy
| | - Désirée Baruffaldi
- Dipartimento di Scienza Applicata e Tecnologia, PolitoBIOMed Lab, Politecnico di Torino, Turin 10129, Italy
| | - Raquel Cue Lopez
- Dipartimento di Scienza Applicata e Tecnologia, PolitoBIOMed Lab, Politecnico di Torino, Turin 10129, Italy
| | - Camilla Paoletti
- Dipartimento di Ingegneria Meccanica e Aerospaziale, Politecnico di Torino, Turin 10129, Italy
| | - Paula Bosch
- Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28006, Spain
| | - Lucia Napione
- Dipartimento di Scienza Applicata e Tecnologia, PolitoBIOMed Lab, Politecnico di Torino, Turin 10129, Italy
| | - Andrea M Giovannozzi
- Quantum Metrology and Nano Technologies Division, National Institute of Metrological Research, Turin 10135, Italy
| | - Candido Fabrizio Pirri
- Dipartimento di Scienza Applicata e Tecnologia, PolitoBIOMed Lab, Politecnico di Torino, Turin 10129, Italy
- Center for Sustainable Futures, Istituto Italiano di Tecnologia, Turin 10144, Italy
| | - Enrique Martinez-Campos
- Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28006, Spain
- Grupo de Síntesis Orgánica y Bioevaluación, Instituto Pluridisciplinar (UCM), Madrid 28040, Spain
| | - Francesca Frascella
- Dipartimento di Scienza Applicata e Tecnologia, PolitoBIOMed Lab, Politecnico di Torino, Turin 10129, Italy
| |
Collapse
|
18
|
Zheng Y, Zhao J, Nie X, Chitrakar B, Gao J, Sang Y. Mutual adhesion of Lactobacillus spp. to intestinal cells: A review of perspectives on surface layer proteins and cell surface receptors. Int J Biol Macromol 2024; 282:137031. [PMID: 39476894 DOI: 10.1016/j.ijbiomac.2024.137031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/22/2024] [Accepted: 10/27/2024] [Indexed: 11/10/2024]
Abstract
The bacterial ability to adhere and colonize in the gut is a key prerequisite to become a probiotic. Lactobacillus spp. surface layer proteins (SLPs) play an important role for such functions in the human body. Interestingly, all SLPs in spite of their structural variation promote adhesion and colonization. A clear understanding about the binding sites of SLPs with the host and their binding modes would help to precisely reveal the process of Lactobacillus spp.-host interaction. Therefore, in this paper, we have sorted out the Lactobacillus spp. SLPs and their adhesion sites in human intestinal cells. Such SLPs included surface layer protein, motif proteins, binding proteins and moonlighting proteins, while enterocyte adhesion receptors included transmembrane glycoproteins and extracellular matrix proteins. We also summarized the tools to assess the adhesion by Lactobacillus spp. Finally, we recommended that three-dimensional cell models and intestinal microarrays could be major tools for assessing adhesion in the future.
Collapse
Affiliation(s)
- Yixin Zheng
- Hebei Agricultural University, No.2596, Lekai South Street, Baoding, Hebei 86-071000, China
| | - Jinrong Zhao
- Hebei Agricultural University, No.2596, Lekai South Street, Baoding, Hebei 86-071000, China
| | - Xinyu Nie
- Hebei Agricultural University, No.2596, Lekai South Street, Baoding, Hebei 86-071000, China
| | - Bimal Chitrakar
- Hebei Agricultural University, No.2596, Lekai South Street, Baoding, Hebei 86-071000, China
| | - Jie Gao
- Hebei Agricultural University, No.2596, Lekai South Street, Baoding, Hebei 86-071000, China.
| | - Yaxin Sang
- Hebei Agricultural University, No.2596, Lekai South Street, Baoding, Hebei 86-071000, China
| |
Collapse
|
19
|
Birjandi AA, Lynham S, Matalova E, Ghuman M, Sharpe P. Regenerative Potential of PDL-Derived Small Extracellular Vesicles. J Periodontal Res 2024. [PMID: 39581762 DOI: 10.1111/jre.13356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 10/03/2024] [Accepted: 10/06/2024] [Indexed: 11/26/2024]
Abstract
Treatment of gingival fibroblasts with PDL extracellular vesicles results in promotion of Wnt signalling pathway and osteogenic differentiation. PDL secretome shows selective wound healing and matrix remodelling which can have implications for future periodontal regenerative strategies.
Collapse
Affiliation(s)
- Anahid A Birjandi
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, Kings College London, London, UK
| | - Steven Lynham
- Centre of Excellence for Mass Spectrometry, Denmark Hill, King's College London, London, UK
| | - Eva Matalova
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
| | - Mandeep Ghuman
- Centre for Host-Microbiome Interactions. Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - Paul Sharpe
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, Kings College London, London, UK
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
| |
Collapse
|
20
|
Nowitzke J, Bista S, Raman S, Dahal N, Stirnemann G, Popa I. Mechanical Unfolding of Network Nodes Drives the Stress Response of Protein-Based Materials. ACS NANO 2024; 18:31031-31043. [PMID: 39487800 DOI: 10.1021/acsnano.4c07352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2024]
Abstract
Biomaterials synthesized from cross-linked folded proteins have untapped potential for biocompatible, resilient, and responsive implementations, but face challenges due to costly molecular refinement and limited understanding of their mechanical response. Under a stress vector, these materials combine the gel-like response of cross-linked networks with the mechanical unfolding and extension of proteins from well-defined 3D structures to unstructured polypeptides. Yet the nanoscale dynamics governing their viscoelastic response remains poorly understood. This lack of understanding is further exacerbated by the fact that the mechanical stability of protein domains depends not only on their structure, but also on the direction of the force vector. To this end, here we propose a coarse-grained network model based on the physical characteristics of polyproteins and combine it with the mechanical unfolding response of protein domains, obtained from single molecule measurements and steered molecular dynamics simulations, to explain the macroscopic response of protein-based materials to a stress vector. We find that domains are about 10-fold more stable when force is applied along their end-to-end coordinate than along the other tethering geometries that are possible inside the biomaterial. As such, the macroscopic response of protein-based materials is mainly driven by the unfolding of the node-domains and rearrangement of these nodes inside the material. The predictions from our models are then confirmed experimentally using force-clamp rheometry. This model is a critical step toward developing protein-based materials with predictable response and that can enable applications for shape memory and energy storage and dissipation.
Collapse
Affiliation(s)
- Joel Nowitzke
- Department of Physics, University of Wisconsin-Milwaukee, 3135 N Maryland Avenue, Milwaukee, Wisconsin 53211, United States
| | - Sanam Bista
- Department of Physics, University of Wisconsin-Milwaukee, 3135 N Maryland Avenue, Milwaukee, Wisconsin 53211, United States
| | - Sadia Raman
- Department of Physics, University of Wisconsin-Milwaukee, 3135 N Maryland Avenue, Milwaukee, Wisconsin 53211, United States
| | - Narayan Dahal
- Department of Physics, University of Wisconsin-Milwaukee, 3135 N Maryland Avenue, Milwaukee, Wisconsin 53211, United States
| | - Guillaume Stirnemann
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne University, CNRS, Paris 75005, France
| | - Ionel Popa
- Department of Physics, University of Wisconsin-Milwaukee, 3135 N Maryland Avenue, Milwaukee, Wisconsin 53211, United States
| |
Collapse
|
21
|
Zeyer KA, Bornert O, Nelea V, Bao X, Leytens A, Sharoyan S, Sengle G, Antonyan A, Bruckner-Tuderman L, Dengjel J, Reinhardt DP, Nyström A. Dipeptidyl Peptidase-4-Mediated Fibronectin Processing Evokes a Profibrotic Extracellular Matrix. J Invest Dermatol 2024; 144:2477-2487.e13. [PMID: 38570029 DOI: 10.1016/j.jid.2024.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/21/2024] [Accepted: 03/13/2024] [Indexed: 04/05/2024]
Abstract
Fibronectin serves as a platform to guide and facilitate deposition of collagen and fibrillin microfibrils. During development of fibrotic diseases, altered fibronectin deposition in the extracellular matrix (ECM) is generally an early event. After this, dysregulated organization of fibrillins and fibrillar collagens occurs. Because fibronectin is an essential orchestrator of healthy ECM, perturbation of its ECM-organizational capacity may be involved in development of fibrosis. To investigate this, we employed recessive dystrophic epidermolysis bullosa as a disease model with progressive, severe dermal fibrosis. Fibroblasts from donors with recessive dystrophic epidermolysis bullosa in 2-dimensional and 3-dimensional cultures displayed dysregulated fibronectin deposition. Our analyses revealed that increase of profibrotic dipeptidyl peptidase-4-positive fibroblasts coincides with altered fibronectin deposition. Dipeptidyl peptidase-4 inhibitors normalized deposition of fibronectin and subsequently of fibrillin microfibrils and collagen I. Intriguingly, proteomics and inhibitor and mutagenesis studies disclosed that dipeptidyl peptidase-4 modulates ECM deposition through the proteolysis of the fibronectin N-terminus. Our study provides mechanistic insights into the observed profibrotic activities of dipeptidyl peptidase-4 and extends the understanding of fibronectin-guided ECM assembly in health and disease.
Collapse
Affiliation(s)
- Karina A Zeyer
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany
| | - Olivier Bornert
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany
| | - Valentin Nelea
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Xinyi Bao
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Alexandre Leytens
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Svetlana Sharoyan
- H. Buniatian Institute of Biochemistry of Armenian NAS, Yerevan, Republic of Armenia
| | - Gerhard Sengle
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany; Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Cologne Center for Musculoskeletal Biomechanics (CCMB), Cologne, Germany
| | - Alvard Antonyan
- H. Buniatian Institute of Biochemistry of Armenian NAS, Yerevan, Republic of Armenia
| | - Leena Bruckner-Tuderman
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
22
|
Ferreira AM, Silva-Álvarez V, Kraev I, Uysal-Onganer P, Lange S. Extracellular vesicles and citrullination signatures are novel biomarkers in sturgeon (Acipenser gueldenstaedtii) during chronic stress due to seasonal temperature challenge. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109974. [PMID: 39426640 DOI: 10.1016/j.fsi.2024.109974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/03/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
Acipenser gueldenstaedtii is one of the most cultured sturgeon species worldwide and of considerable economic value for caviar production. There are though considerable challenges around chronic stress responses due to increased summer temperatures, impacting sturgeons' immune responses and their susceptibility to opportunistic infections. The identification of molecular and cellular pathways involved in stress responses may contribute to identifying novel biomarkers reflective of fish health status, crucial for successful sturgeon aquaculture. Protein citrullination is a calcium-catalysed post-translational modification caused by peptidylarginine deiminases (PADs), altering target protein function and affecting protein interactions in physiological and pathobiological processes. PADs can also modulate extracellular vesicle (EVs) profiles, which play critical roles in cellular communication, via transport of their cargoes (proteins, including post-translationally modified proteins, genetic material and micro-RNAs). This study identified differences in EV signatures, and citrullinated proteins in sera from winter and summer farmed sturegeons. EVs were significantly elevated in sera of the summer chronically stressed group. The citrullinated proteins and associated gene ontology (GO) pathways in sera and serum-EVs of chronically heat stressed A. gueldenstaedtii, showed some changes, with specific citrullinated serum protein targets including alpa-2-macroglobulin, alpha globin, calcium-dependent secretion activator, ceruloplasmin, chemokine XC receptor, complement C3 isoforms, complement C9, plectin, selenoprotein and vitellogenin. In serum-EVs, citrullinated protein cargoes identified only in the chronically stressed summer group included alpha-1-antiproteinase, apolipoprotein B-100, microtubule actin crosslinking factor and histone H3. Biological gene ontology (GO) pathways related to citrullinated serum proteins in the chronically stressed group were associated with innate and adaptive immune responses, stress responses and metabolic processes. In serum-EVs of the heat-stressed group the citrullinome associated with various metabolic GO pathways. In addition to modified citrullinated protein content, Serum-EVs from the stressed summer group showed significantly increased levels of the inflammatory associated miR-155 and the hypoxia-associated miR-210, but significantly reduced levels of the growth-associated miR-206. Our findings highlight roles for protein citrullination and EV signatures in response to chronic heat stress in A. gueldenstaedtii, indicating a trade-off in immunity versus growth and may be of value for sturgeon aquaculture.
Collapse
Affiliation(s)
- Ana María Ferreira
- Laboratorio de Inmunología, Instituto de Química Biológica, Facultad de Ciencias, Instituto de Higiene, UdelaR, Uruguay; Área de Inmunología, Departamento de Biociencias, Facultad de Química, Instituto de Higiene, UdelaR, Uruguay.
| | - Valeria Silva-Álvarez
- Laboratorio de Inmunología, Instituto de Química Biológica, Facultad de Ciencias, Instituto de Higiene, UdelaR, Uruguay; Área de Inmunología, Departamento de Biociencias, Facultad de Química, Instituto de Higiene, UdelaR, Uruguay.
| | - Igor Kraev
- Electron Microscopy Suite, Faculty of Science, Technology, Engineering and Mathematics, Open University, Milton Keynes MK7 6AA, UK.
| | - Pinar Uysal-Onganer
- Cancer Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK.
| | - Sigrun Lange
- Pathobiology and Extracellular Vesicles Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK.
| |
Collapse
|
23
|
Velarde K, Arvonen A, Gonzalez T, Diller RB. A Biologic and Physical Characterization of an Injectable Amniotic Membrane Designed for Treating Diabetic Foot Ulcers. Bioengineering (Basel) 2024; 11:1087. [PMID: 39593747 PMCID: PMC11591430 DOI: 10.3390/bioengineering11111087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
INTRODUCTION Globally, the health and quality of life of millions of people are negatively affected by diabetic foot ulcers (DFUs). To treat these chronic wounds, a novel injectable drug for closing DFUs composed of micronized amniotic membrane was developed. This new therapeutic drug for wound repair expands on traditional allograft therapies by allowing extracellular matrix proteins, growth factors, and cytokines to reach wound anatomies in DFUs that are difficult to treat. The aim of this study was to evaluate the components of the injectable drug. METHODS Liquid chromatography with tandem mass spectrometry and a Quantibody® human cytokine array were conducted to identify and characterize growth factors and proteins known to contribute to wound healing. In addition, hyaluronic acid was quantified and compared between the injectable and human amniotic fluid using a hyaluronan enzyme-linked immunosorbent assay. Cell proliferation, migration, angiogenesis, and viability were evaluated to assess the performance of the novel injectable in vitro. The rheometric properties of the product were evaluated by assessing it pre- and post-injection through a 22-gauge needle to measure the viscosity using a shear- and temperature-dependent viscosity protocol. RESULTS Liquid chromatography with tandem mass spectrometry and Quantibody® human cytokine array revealed growth factors and proteins imperative for wound healing. The quantified hyaluronic acid was compared between the injectable and human amniotic fluid, resulting in a statistically significant difference, with higher protein concentrations found in the injectable. In vitro qualitative and quantitative analysis confirmed an increase in cell viability, proliferation, and migration when treated with the drug. An evaluation of the rheometric properties of the injectable drug after passing through a 22-gauge cannula presented no alterations to the biologic drug. CONCLUSIONS Collectively, these data present the potential of a novel injectable drug for the treatment of DFUs.
Collapse
Affiliation(s)
- Kimberly Velarde
- Amnio Technology, LLC., 22510 N. 18th Dr., Phoenix, AZ 85027, USA; (K.V.); (A.A.); (T.G.)
| | - Audrey Arvonen
- Amnio Technology, LLC., 22510 N. 18th Dr., Phoenix, AZ 85027, USA; (K.V.); (A.A.); (T.G.)
| | - Tatyana Gonzalez
- Amnio Technology, LLC., 22510 N. 18th Dr., Phoenix, AZ 85027, USA; (K.V.); (A.A.); (T.G.)
| | - Robert B. Diller
- Amnio Technology, LLC., 22510 N. 18th Dr., Phoenix, AZ 85027, USA; (K.V.); (A.A.); (T.G.)
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ 86011, USA
| |
Collapse
|
24
|
Kempuraj D, Dourvetakis KD, Cohen J, Valladares DS, Joshi RS, Kothuru SP, Anderson T, Chinnappan B, Cheema AK, Klimas NG, Theoharides TC. Neurovascular unit, neuroinflammation and neurodegeneration markers in brain disorders. Front Cell Neurosci 2024; 18:1491952. [PMID: 39526043 PMCID: PMC11544127 DOI: 10.3389/fncel.2024.1491952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Neurovascular unit (NVU) inflammation via activation of glial cells and neuronal damage plays a critical role in neurodegenerative diseases. Though the exact mechanism of disease pathogenesis is not understood, certain biomarkers provide valuable insight into the disease pathogenesis, severity, progression and therapeutic efficacy. These markers can be used to assess pathophysiological status of brain cells including neurons, astrocytes, microglia, oligodendrocytes, specialized microvascular endothelial cells, pericytes, NVU, and blood-brain barrier (BBB) disruption. Damage or derangements in tight junction (TJ), adherens junction (AdJ), and gap junction (GJ) components of the BBB lead to increased permeability and neuroinflammation in various brain disorders including neurodegenerative disorders. Thus, neuroinflammatory markers can be evaluated in blood, cerebrospinal fluid (CSF), or brain tissues to determine neurological disease severity, progression, and therapeutic responsiveness. Chronic inflammation is common in age-related neurodegenerative disorders including Alzheimer's disease (AD), Parkinson's disease (PD), and dementia. Neurotrauma/traumatic brain injury (TBI) also leads to acute and chronic neuroinflammatory responses. The expression of some markers may also be altered many years or even decades before the onset of neurodegenerative disorders. In this review, we discuss markers of neuroinflammation, and neurodegeneration associated with acute and chronic brain disorders, especially those associated with neurovascular pathologies. These biomarkers can be evaluated in CSF, or brain tissues. Neurofilament light (NfL), ubiquitin C-terminal hydrolase-L1 (UCHL1), glial fibrillary acidic protein (GFAP), Ionized calcium-binding adaptor molecule 1 (Iba-1), transmembrane protein 119 (TMEM119), aquaporin, endothelin-1, and platelet-derived growth factor receptor beta (PDGFRβ) are some important neuroinflammatory markers. Recent BBB-on-a-chip modeling offers promising potential for providing an in-depth understanding of brain disorders and neurotherapeutics. Integration of these markers in clinical practice could potentially enhance early diagnosis, monitor disease progression, and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Kirk D. Dourvetakis
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Jessica Cohen
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Daniel Seth Valladares
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Rhitik Samir Joshi
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Sai Puneeth Kothuru
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
- College of Psychology, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Tristin Anderson
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Baskaran Chinnappan
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Amanpreet K. Cheema
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Nancy G. Klimas
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
- Miami VA Geriatric Research Education and Clinical Center (GRECC), Miami Veterans Affairs Healthcare System, Miami, FL, United States
| | - Theoharis C. Theoharides
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
- Department of Immunology, Tufts, University School of Medicine, Boston, MA, United States
| |
Collapse
|
25
|
Di T, Wang L, Cheng B, Guo M, Feng C, Wu Z, Wang L, Chen Y. Single-cell RNA sequencing reveals vascularization-associated cell subpopulations in dental pulp: PDGFRβ+ DPSCs with activated PI3K/AKT pathway. Stem Cells 2024; 42:914-927. [PMID: 39167061 DOI: 10.1093/stmcls/sxae051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND This study aims to address challenges in dental pulp regeneration therapy. The heterogeneity of DPSCs poses challenges, especially in stem cell transplantation for clinical use, particularly when sourced from donors of different ages and conditions. METHODS Pseudotime analysis was employed to analyze single-cell sequencing data, and immunohistochemical studies were conducted to investigate the expression of fibronectin 1 (FN1). We performed in vitro sorting of PDGFRβ+ DPSCs using flow cytometry. A series of functional assays, including cell proliferation, scratch, and tube formation assays, were performed to experimentally validate the vasculogenic capabilities of the identified PDGFRβ+ DPSC subset. Furthermore, gene-edited mouse models were utilized to demonstrate the importance of PDGFRβ+ DPSCs. Transcriptomic sequencing was conducted to compare the differences between PDGFRβ+ DPSCs and P1-DPSCs. RESULTS Single-cell sequencing analysis unveiled a distinct subset, PDGFRβ+ DPSCs, characterized by significantly elevated FN1 expression during dental pulp development. Subsequent cell experiments demonstrated that this subset possesses remarkable abilities to promote HUVEC proliferation, migration, and tube formation. Gene-edited mouse models confirmed the vital role of PDGFRβ+ DPSCs in dental pulp development. Transcriptomic sequencing and in vitro experiments demonstrated that the PDGFR/PI3K/AKT signaling pathway is a crucial factor mediating the proliferation rate and pro-angiogenic properties of PDGFRβ+ DPSCs. CONCLUSION We defined a new subset, PDGFRβ+ DPSCs, characterized by strong proliferative activity and pro-angiogenic capabilities, demonstrating significant clinical translational potential.
Collapse
Affiliation(s)
- Tiankai Di
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
- Department of Stomatology, Joint Logistics Support Force of the Chinese People's Liberation Army, Hohhot, Inner Mongolia 010000, People's Republic of China
| | - Liying Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Baixiang Cheng
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Department of General Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Mingzhu Guo
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao 266001, Shandong Province, People's Republic of China
| | - Chao Feng
- Department of Clinical Laboratory, Joint Logistics Support Force of the Chinese People's Liberation Army, Hohhot, Inner Mongolia 010000, People's Republic of China
| | - Zhenzhen Wu
- Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region of China, People's Republic of China
| | - Lulu Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Yujiang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| |
Collapse
|
26
|
Yu S, Wang S, Wang X, Xu X. The axis of tumor-associated macrophages, extracellular matrix proteins, and cancer-associated fibroblasts in oncogenesis. Cancer Cell Int 2024; 24:335. [PMID: 39375726 PMCID: PMC11459962 DOI: 10.1186/s12935-024-03518-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 09/29/2024] [Indexed: 10/09/2024] Open
Abstract
The extracellular matrix (ECM) is a complex, dynamic network of multiple macromolecules that serve as a crucial structural and physical scaffold for neighboring cells. In the tumor microenvironment (TME), ECM proteins play a significant role in mediating cellular communication between cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs). Revealing the ECM modification of the TME necessitates the intricate signaling cascades that transpire among diverse cell populations and ECM proteins. The advent of single-cell sequencing has enabled the identification and refinement of specific cellular subpopulations, which has substantially enhanced our comprehension of the intricate milieu and given us a high-resolution perspective on the diversity of ECM proteins. However, it is essential to integrate single-cell data and establish a coherent framework. In this regard, we present a comprehensive review of the relationships among ECM, TAMs, and CAFs. This encompasses insights into the ECM proteins released by TAMs and CAFs, signaling integration in the TAM-ECM-CAF axis, and the potential applications and limitations of targeted therapies for CAFs. This review serves as a reliable resource for focused therapeutic strategies while highlighting the crucial role of ECM proteins as intermediates in the TME.
Collapse
Affiliation(s)
- Shuhong Yu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Siyu Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xuanyu Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ximing Xu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
27
|
Han Y, Liu C, Chen S, Sun H, Jia Z, Shi J, Wang L, Du K, Chang Y. Columbianadin ameliorates rheumatoid arthritis by attenuating synoviocyte hyperplasia through targeted vimentin to inhibit the VAV2/Rac-1 signaling pathway. J Adv Res 2024:S2090-1232(24)00432-6. [PMID: 39369957 DOI: 10.1016/j.jare.2024.09.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/13/2024] [Accepted: 09/29/2024] [Indexed: 10/08/2024] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is an autoimmune disease pathologically characterized by synovial inflammation. The abnormal activation of synoviocytes seems to accompany the progression of RA. The role and exact molecular mechanism in RA of columbianadin (CBN) which is a natural coumarin is still unclear. OBJECTIVES The present research aimed to investigate the effect of vimentin on the abnormal growth characteristics of RA synoviocytes and the targeted regulatory role of CBN. METHODS Cell migration and invasion were detected using the wound healing and transwell method. Mechanistically, the direct molecular targets of CBN were screened and identified by activity-based protein profiling. The expression of relevant proteins and mRNA in cells and mouse synovium was detected by western blotting and qRT-PCR. Changes in the degree of paw swelling and body weight of mice were recorded. H&E staining, toluidine blue staining, and micro-CT were used to visualize the degree of pathological damage in the ankle joints of mice. Small interfering RNA and plasmid overexpression of vimentin were used to observe their effects on MH7A cell proliferation, migration, apoptosis, and downstream molecular signaling. RESULTS The TNF-α-induced proliferation and migration of MH7A cells could be significantly repressed by CBN (25,50 μM), and the expression of apoptosis and autophagy-associated proteins could be modulated. Furthermore, CBN could directly bind to vimentin and inhibit its expression and function in synoviocytes, thereby ameliorating foot and paw swelling and joint damage in CIA mice. Silencing and overexpression of vimentin might be involved in developing RA synovial hyperplasia and invasive cartilage by activating VAV2 phosphorylation-mediated expression of Rac-1, which affects abnormal growth characteristics, such as synoviocyte invasion and migration. CONCLUSION CBN-targeted vimentin restrains the overactivation of RA synoviocytes thereby delaying the pathological process in CIA mice, which provides valuable targets and insights for understanding the pathological mechanisms of RA synovial hyperplasia.
Collapse
Affiliation(s)
- Yuli Han
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Changqing Liu
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shujing Chen
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Huihui Sun
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Zhaoyu Jia
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jiaxin Shi
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lirong Wang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Kunze Du
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yanxu Chang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
28
|
Zhang Y, Guo Y, Song L, Liu W, Nian R, Fan X. Streamlined on-column refolding and purification of nanobodies from inclusion bodies expressed as fusion proteins. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1246:124279. [PMID: 39186888 DOI: 10.1016/j.jchromb.2024.124279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 08/14/2024] [Accepted: 08/18/2024] [Indexed: 08/28/2024]
Abstract
This study introduces an efficient on-column refolding and purification method for preparing nanobodies (Nbs) expressed as inclusion bodies and fusion proteins. The HisTrapTM FF system was successfully employed for the purification of the fusion protein FN1-ΔI-CM-2D5. The intein ΔI-CM cleavage activity was activated at 42 °C, followed by incubation for 4 h. Leveraging the remarkable thermal stability of Nbs, 2D5 was further purified through heat treatment at 80 °C for 1h. This method yielded up to 107.2 mg of pure 2D5 with a purity of 99.2 % from just 1L of bacterial culture grown in a shaker flask. Furthermore, this approach successfully restored native secondary structure and affinity of 2D5. Additionally, the platform was effectively applied to the refolding and purification of a polystyrene-binding nanobody (B2), which exhibited limited expression in the periplasmic and cytoplasmic spaces of E. coli. This endeavor resulted in the isolation of 53.2 mg of pure B2 Nb with a purity exceeding 99.5 % from the same volume of bacterial culture. Significantly, this approach restored the native secondary structure of the Nbs, highlighting its potential for addressing challenges associated with expressing complex Nbs in E. coli. Overall, this innovative platform provides a scientifically rigorous and reproducible method for the efficient preparation of Nbs, offering a valuable tool for antibody research and development.
Collapse
Affiliation(s)
- Yiwen Zhang
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China; University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing 100049, China
| | - Yang Guo
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China; University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Beijing 100049, China
| | - Liang Song
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China
| | - Wenshuai Liu
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China
| | - Rui Nian
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China
| | - Xiying Fan
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, No. 189, Songling Road, Qingdao 266101, China; Shandong Energy Institute, No. 189, Songling Road, Qingdao 266101, China; Qingdao New Energy Shandong Laboratory, No. 189, Songling Road, Qingdao 266101, China.
| |
Collapse
|
29
|
Kim SH, Shin HL, Son TH, Lim SA, Kim D, Yoon JH, Choi H, Kim HG, Choi SW. Quercus glauca Acorn Seed Coat Extract Promotes Wound Re-Epithelialization by Facilitating Fibroblast Migration and Inhibiting Dermal Inflammation. BIOLOGY 2024; 13:775. [PMID: 39452084 PMCID: PMC11505045 DOI: 10.3390/biology13100775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
The skin, recognized as the largest organ in the human body, serves a vital function in safeguarding against external threats. Severe damage to the skin can pose significant risks to human health. There is an urgent requirement for safe and effective therapies for wound healing. While phytotherapy has been widely utilized for various health conditions, the potential of Quercus glauca in promoting wound healing has not been thoroughly explored. Q. glauca is a cultivated crop known for its abundance of bioactive compounds. This study examined the wound-healing properties of Quercus glauca acorn seed coat water extract (QGASE). The findings from the study suggest that QGASE promotes wound closure in HF cells by upregulating essential markers related to the wound-healing process. Additionally, QGASE demonstrates antioxidant effects, mitigating oxidative stress and aiding in recovery from injuries induced by H2O2. In vivo experiments provide additional substantiation supporting the efficacy of QGASE in enhancing wound healing. The collective results indicate that QGASE may be a promising candidate for the development of innovative therapeutic strategies aimed at enhancing skin wound repair.
Collapse
Affiliation(s)
- Shin-Hye Kim
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Jinju 52817, Republic of Korea; (S.-H.K.); (H.-L.S.); (T.H.S.); (D.K.); (J.-H.Y.)
| | - Hye-Lim Shin
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Jinju 52817, Republic of Korea; (S.-H.K.); (H.-L.S.); (T.H.S.); (D.K.); (J.-H.Y.)
- Department of Biological Sciences, Jeonbuk National University, Jeonju 54896, Republic of Korea;
| | - Tae Hyun Son
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Jinju 52817, Republic of Korea; (S.-H.K.); (H.-L.S.); (T.H.S.); (D.K.); (J.-H.Y.)
| | - So-An Lim
- Pharmacogenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea;
| | - Dongsoo Kim
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Jinju 52817, Republic of Korea; (S.-H.K.); (H.-L.S.); (T.H.S.); (D.K.); (J.-H.Y.)
| | - Jun-Hyuck Yoon
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Jinju 52817, Republic of Korea; (S.-H.K.); (H.-L.S.); (T.H.S.); (D.K.); (J.-H.Y.)
| | - Hyunmo Choi
- Department of Forest Bioresources, National Institute of Forest Science (NIFoS), Suwon 16631, Republic of Korea;
| | - Hwan-Gyu Kim
- Department of Biological Sciences, Jeonbuk National University, Jeonju 54896, Republic of Korea;
| | - Sik-Won Choi
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Jinju 52817, Republic of Korea; (S.-H.K.); (H.-L.S.); (T.H.S.); (D.K.); (J.-H.Y.)
| |
Collapse
|
30
|
Patten J, Halligan P, Bashiri G, Kegel M, Bonadio JD, Wang K. EDA Fibronectin Microarchitecture and YAP Translocation During Wound Closure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614581. [PMID: 39386582 PMCID: PMC11463502 DOI: 10.1101/2024.09.23.614581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Fibronectin (Fn) is an extracellular matrix glycoprotein with mechanosensitive structure-function. EDA Fn, a Fn isoform, is not present in adult tissue but is required for tissue repair. Curiously, EDA Fn is linked to both regenerative and fibrotic tissue repair. Given that Fn mechanoregulates cell behavior, Fn EDA organization during wound closure might play a role in mediating these differing responses. One mechanism by which cells sense and respond to their microenvironment is by activating a transcriptional co-activator, Yes-associated protein (YAP). Interestingly, YAP activity is not only required for wound closure, but similarly linked to both regenerative and fibrotic repair. Therefore, this study aims to evaluate how, during normal and fibrotic wound closure, EDA Fn organization might modulate YAP translocation by culturing human dermal fibroblasts on polydimethylsiloxane (PDMS) substrates mimicking normal (soft: 18 kPa) and fibrotic (stiff: 146 kPa) wounded skin. On stiffer substrates mimicking fibrotic wounds, fibroblasts assembled an aligned EDA Fn matrix comprising thinner fibers, suggesting increased microenvironmental tension. To evaluate if cell binding to the EDA domain of Fn was essential to overall matrix organization, fibroblasts were treated with Irigenin, which inhibits binding to the EDA domain within Fn. Blocking adhesion to EDA led to randomly organized EDA Fn matrices with thicker fibers, suggesting reduced microenvironmental tension even during fibrotic wound closure. To evaluate if YAP signaling plays a role in EDA Fn organization, fibroblasts were treated with CA3, which suppresses YAP activity in a dose-dependent manner. Treatment with CA3 also led to randomly organized EDA Fn matrices with thicker fibers, suggesting a potential connected mechanism of reducing tension during fibrotic wound closure. Next, YAP activity was assessed to evaluate the impact of EDA Fn organization. Interestingly, fibroblasts migrating on softer substrates mimicking normal wounds increased YAP activity but on stiffer substrates, decreased YAP activity. When fibroblasts on stiffer substrates were treated with Irigenin or CA3, fibroblasts increased YAP activity. These results suggest there may be disrupted signaling between EDA Fn organization and YAP translocation during fibrotic wound closure that could be restored when reestablishing normal EDA Fn matrix organization to instead drive regenerative wound repair.
Collapse
Affiliation(s)
- Jennifer Patten
- Department of Bioengineering, Temple University, Pennsylvania
| | | | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Pennsylvania
| | - Michael Kegel
- Department of Bioengineering, Temple University, Pennsylvania
| | - Jacob D Bonadio
- Department of Bioengineering, Temple University, Pennsylvania
| | - Karin Wang
- Department of Bioengineering, Temple University, Pennsylvania
| |
Collapse
|
31
|
Min J, Jiaqi H, Lihua L, Qianqian C, Shujuan W, Xiang L, Liang L, Liang R, Yiwu Z, Qian L. Proteomics of severe SARS-COV-2 infection and paraquat poisoning in human lung tissue samples: comparison of microbial infected and toxic pulmonary fibrosis. Front Cell Infect Microbiol 2024; 14:1446305. [PMID: 39301288 PMCID: PMC11410708 DOI: 10.3389/fcimb.2024.1446305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 08/12/2024] [Indexed: 09/22/2024] Open
Abstract
Introduction Pulmonary fibrosis (PF) encompasses a spectrum of lung conditions characterized by the abnormal accumulation of scar tissue in the lungs, leading to impaired respiratory function. Various conditions can result in severe PF, among which viral infections have emerged as significant triggers. In addition to viral infections, exposure to toxic substances such as paraquat represents another significant risk factor for PF. Therefore, this study aimed to explore the dissimilarities and similarities between PF triggered by viral infections and chemical toxicants, using the mechanism of PF in IPF as a reference. Methods Data-independent acquisition proteomics technology was employed to identify COVID-19 and paraquat-induced PF from the autopsy of lung tissue samples obtained from individuals who died due to PF. Bioinformatics was employed for differential protein analysis, and selected indicators were validated on pathological sections. Results Our results showed that the differential proteins associated with the two causes of PF were enriched in similar lung fibrosis-related signaling pathways, such as the Wnt signaling pathway. However, differences were observed in proteins such as CACYBP, we verified the consistency of the results with proteomics using the IHC approach. Conclusion This study illuminates distinct protein-level differences by investigating pulmonary fibrosis pathways in severe COVID-19 and paraquat poisoning. Although both conditions activate lung-protective and repair pathways, COVID-19 shows limited phosphorylation-independent ubiquitination of β-catenin compared to paraquat toxicity. These findings shed light on potential therapeutic targets for PF induced via diverse factors.
Collapse
Affiliation(s)
- Jiang Min
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hou Jiaqi
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Lihua
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chai Qianqian
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wang Shujuan
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liu Xiang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liu Liang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ren Liang
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhou Yiwu
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liu Qian
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
32
|
Ahmad N, Kiriako G, Saliba J, Abla K, El-Sabban M, Mhanna R. Engineering a 3D Biomimetic Peptides Functionalized-Polyethylene Glycol Hydrogel Model Cocultured with Endothelial Cells and Astrocytes: Enhancing In Vitro Blood-Brain Barrier Biomimicry. Mol Pharm 2024; 21:4664-4672. [PMID: 39133897 PMCID: PMC11372828 DOI: 10.1021/acs.molpharmaceut.4c00599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/03/2024]
Abstract
The blood-brain barrier (BBB) poses a significant challenge for drug delivery and is linked to various neurovascular disorders. In vitro BBB models provide a tool to investigate drug permeation across the BBB and the barrier's response to external injury events. Yet, existing models lack fidelity in replicating the BBB's complexity, hindering a comprehensive understanding of its functions. This study introduces a three-dimensional (3D) model using polyethylene glycol (PEG) hydrogels modified with biomimetic peptides that represent recognition sequences of key proteins in the brain. Hydrogels were functionalized with recognition sequences for laminin (IKVAV) and fibronectin peptides (RGD) and chemically cross-linked with matrix metalloprotease-sensitive peptides (MMPs) to mimic the extracellular matrix of the BBB. Astrocytes and endothelial cells were seeded within and on the surface of the hydrogels, respectively. The barrier integrity was assessed through different tests including transendothelial electrical resistance (TEER), the permeability of sodium fluorescence (Na-F), the permeability of Evan's blue bound to albumin (EBA), and the expression of zonula occluden-1 (ZO-1) in seeded endothelial cells. Hydrogels with a combination of RGD and IKVAV peptides displayed superior performance, exhibiting significantly higher TEER values (55.33 ± 1.47 Ω·cm2) at day 5 compared to other 2D controls including HAECs-monoculture and HAECs-cocultured with NHAs seeded on well inserts and 3D controls including RGD hydrogel and RGD-IKVAV monoculture with HAECs and RGD hydrogel cocultured with HAECs and NHAs. The designed 3D system resulted in the lowest Evan's blue permeability at 120 min (0.215 ± 0.055 μg/mL) compared to controls. ZO-1 expression was significantly higher and formed a relatively larger network in the functionalized hydrogel cocultured with astrocytes and endothelial cells compared to the controls. Thus, the designed 3D model effectively recapitulates the main BBB structure and function in vitro and is expected to contribute to a deeper understanding of pathological CNS angiogenesis and the development of effective CNS medications.
Collapse
Affiliation(s)
- Nesrine Ahmad
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Georges Kiriako
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107-2020, Lebanon
| | - John Saliba
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107-2020, Lebanon
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Kawthar Abla
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Rami Mhanna
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107-2020, Lebanon
| |
Collapse
|
33
|
Guo M, Li Y, Tang J, Wang Q, Wang Q, Zhou H, Lin H, Ma Z, Fan H. Glaesserella parasuis serotype 4 exploits fibronectin via RlpA for tracheal colonization following porcine circovirus type 2 infection. PLoS Pathog 2024; 20:e1012513. [PMID: 39264911 PMCID: PMC11392263 DOI: 10.1371/journal.ppat.1012513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/16/2024] [Indexed: 09/14/2024] Open
Abstract
Porcine circovirus type 2 (PCV2) often causes disease through coinfection with other bacterial pathogens, including Glaesserella parasuis (G. parasuis), which causes high morbidity and mortality, but the role played by PCV2 and bacterial and host factors contributing to this process have not been defined. Bacterial attachment is assumed to occur via specific receptor-ligand interactions between adhesins on the bacterial cell and host proteins adsorbed to the implant surface. Mass spectrometry (MS) analysis of PCV2-infected swine tracheal epithelial cells (STEC) revealed that the expression of Extracellular matrix protein (ECM) Fibronectin (Fn) increased significantly on the infected cells surface. Importantly, efficient G. parasuis serotype 4 (GPS4) adherence to STECs was imparted by interactions with Fn. Furthermore, abrogation of adherence was gained by genetic knockout of Fn, Fn and Integrin β1 antibody blocking. Fn is frequently exploited as a receptor for bacterial pathogens. To explore the GPS4 adhesin that interacts with Fn, recombinant Fn N-terminal type I and type II domains were incubated with GPS4, and the interacting proteins were pulled down for MS analysis. Here, we show that rare lipoprotein A (RlpA) directly interacts with host Fibronectin mediating GPS4 adhesion. Finally, we found that PCV2-induced Fibronectin expression and adherence of GPS4 were prevented significantly by TGF-β signaling pathway inhibitor SB431542. Our data suggest the RlpA-Fn interaction to be a potentially promising novel therapeutic target to combat PCV2 and GPS4 coinfection.
Collapse
Affiliation(s)
- Mengru Guo
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yuhui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jinsheng Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Qing Wang
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Qiancheng Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hong Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Huixing Lin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhe Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hongjie Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- College of Animal Science, Anhui Science and Technology University, Fengyang, China
| |
Collapse
|
34
|
Yan Y, Yan Q, Cai K, Wang Z, Li Q, Zhao K, Jian Y, Jia X. Silk fibroin microgrooved zirconia surfaces improve connective tissue sealing through mediating glycolysis of fibroblasts. Mater Today Bio 2024; 27:101158. [PMID: 39081464 PMCID: PMC11287005 DOI: 10.1016/j.mtbio.2024.101158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 08/02/2024] Open
Abstract
The use of zirconia has significantly enhanced the aesthetic outcomes of implant restorations. However, peri-implantitis remains a challenge to long-term functionality of implants. Unlike the perpendicularly arranged collagen fibers in periodontal tissue, those in peri-implant tissue lie parallel to the abutment surface and contain fewer fibroblasts, making them more prone to inflammation. Studies have shown that microgroove structures on implant abutments could improve surrounding soft tissue structure. However, creating precise microgrooves on zirconia without compromising its mechanical integrity is technically challenging. In this study, we applied inkjet printing, an additive manufacturing technique, to create stable silk fibroin microgroove (SFMG) coatings of various dimensions on zirconia substrates. SFMG significantly improved the hydrophilicity of zirconia and showed good physical and chemical stability. The SFMG with 90 μm interval and 10 μm depth was optimal in promoting the proliferation, alignment, and extracellular matrix production of human gingival fibroblasts (HGFs). Moreover, the in vitro results revealed that SFMG stimulated key glycolytic enzyme gene expression in HGFs via the PI3K-AKT-mTOR pathway. Additionally, the in vivo results of histological staining of peri-abutments soft tissue showed that SFMG promoted the vertical alignment of collagen fibers relative to the abutment surface, improving connective tissue sealing around the zirconia abutment. Our results indicated that SFMG on zirconia can enhance HGF proliferation, migration and collagen synthesis by regulating glycolysis though PI3K-AKT-mTor pathway, thereby improving connective tissue sealing.
Collapse
Affiliation(s)
- Yinuo Yan
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuan West Road, Guangzhou, Guangdong, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Qiqian Yan
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuan West Road, Guangzhou, Guangdong, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Kexin Cai
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuan West Road, Guangzhou, Guangdong, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Zhihan Wang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuan West Road, Guangzhou, Guangdong, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Qiulan Li
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuan West Road, Guangzhou, Guangdong, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Ke Zhao
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuan West Road, Guangzhou, Guangdong, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Yutao Jian
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuan West Road, Guangzhou, Guangdong, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Xiaoshi Jia
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuan West Road, Guangzhou, Guangdong, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| |
Collapse
|
35
|
Sirois JP, Heinz A. Matrikines in the skin: Origin, effects, and therapeutic potential. Pharmacol Ther 2024; 260:108682. [PMID: 38917886 DOI: 10.1016/j.pharmthera.2024.108682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/31/2024] [Accepted: 06/21/2024] [Indexed: 06/27/2024]
Abstract
The extracellular matrix (ECM) represents a complex multi-component environment that has a decisive influence on the biomechanical properties of tissues and organs. Depending on the tissue, ECM components are subject to a homeostasis of synthesis and degradation, a subtle interplay that is influenced by external factors and the intrinsic aging process and is often disturbed in pathologies. Upon proteolytic cleavage of ECM proteins, small bioactive peptides termed matrikines can be formed. These bioactive peptides play a crucial role in cell signaling and contribute to the dynamic regulation of both physiological and pathological processes such as tissue remodeling and repair as well as inflammatory responses. In the skin, matrikines exert an influence for instance on cell adhesion, migration, and proliferation as well as vasodilation, angiogenesis and protein expression. Due to their manifold functions, matrikines represent promising leads for developing new therapeutic options for the treatment of skin diseases. This review article gives a comprehensive overview on matrikines in the skin, including their origin in the dermal ECM, their biological effects and therapeutic potential for the treatment of skin pathologies such as melanoma, chronic wounds and inflammatory skin diseases or for their use in anti-aging cosmeceuticals.
Collapse
Affiliation(s)
- Jonathan P Sirois
- Department of Pharmacy, LEO Foundation Center for Cutaneous Drug Delivery, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Heinz
- Department of Pharmacy, LEO Foundation Center for Cutaneous Drug Delivery, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
36
|
Yan L, Wang Y, Feng J, Ni Y, Zhang T, Cao Y, Zhou M, Zhao C. Mechanism and application of fibrous proteins in diabetic wound healing: a literature review. Front Endocrinol (Lausanne) 2024; 15:1430543. [PMID: 39129915 PMCID: PMC11309995 DOI: 10.3389/fendo.2024.1430543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024] Open
Abstract
Diabetic wounds are more complex than normal chronic wounds because of factors such as hypoxia, reduced local angiogenesis, and prolonged inflammation phase. Fibrous proteins, including collagen, fibrin, laminin, fibronectin, elastin etc., possess excellent inherent properties that make them highly advantageous in the area of wound healing. Accumulating evidence suggests that they contribute to the healing process of diabetic wounds by facilitating the repair and remodel of extracellular matrix, stimulating the development of vascular and granulation tissue, and so on. However, there is currently a lack of a comprehensive review of the application of these proteins in diabetes wounds. An overview of fibrous protein characteristics and the alterations linked to diabetic wounds is given in this article's initial section. Next is a summary of the advanced applications of fibrous proteins in the last five years, including acellular dermal matrix, hydrogel, foam, scaffold, and electrospun nanofibrous membrane. These dressings have the ability to actively promote healing in addition to just covering wounds compared to traditional wound dressings like gauze or bandage. Research on fibrous proteins and their role in diabetic wound healing may result in novel therapeutic modalities that lower the incidence of diabetic wounds and thereby enhance the health of diabetic patients.
Collapse
Affiliation(s)
- Lilin Yan
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqing Wang
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiawei Feng
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Ni
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ting Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yemin Cao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingmei Zhou
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cheng Zhao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
37
|
Wang L, He L, Yi W, Wang M, Xu F, Liu H, Nie J, Pan YH, Dang S, Zhang W. ADAMTS18-fibronectin interaction regulates the morphology of liver sinusoidal endothelial cells. iScience 2024; 27:110273. [PMID: 39040056 PMCID: PMC11261151 DOI: 10.1016/j.isci.2024.110273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/12/2024] [Accepted: 06/12/2024] [Indexed: 07/24/2024] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) have a unique morphological structure known as "fenestra" that plays a crucial role in liver substance exchange and homeostasis maintenance. In this study, we demonstrate that ADAMTS18 protease is primarily secreted by fetal liver endothelial cells. ADAMTS18 deficiency leads to enlarged fenestrae and increased porosity of LSECs, microthrombus formation in liver vessels, and an imbalance of liver oxidative stress. These defects worsen carbon tetrachloride (CCl4)-induced liver fibrosis and diethylnitrosamine (DEN)/high-fat-induced hepatocellular carcinoma (HCC) in adult Adamts18-deficient mice. Mechanically, ADAMTS18 functions as a modifier of fibronectin (FN) to regulate the morphological acquisition of LSECs via the vascular endothelial growth factor A (VEGFA) signaling pathways. Collectively, a mechanism is proposed for LSEC morphogenesis and liver homeostasis maintenance via ADAMTS18-FN-VEGFA niches.
Collapse
Affiliation(s)
- Liya Wang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Li He
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Weijia Yi
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Min Wang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Fangmin Xu
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Hanlin Liu
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiahui Nie
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Yi-Hsuan Pan
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| | - Suying Dang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
38
|
Liang NE, Parker JB, Lu JM, Januszyk M, Wan DC, Griffin M, Longaker MT. Understanding the Foreign Body Response via Single-Cell Meta-Analysis. BIOLOGY 2024; 13:540. [PMID: 39056733 PMCID: PMC11273435 DOI: 10.3390/biology13070540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Foreign body response (FBR) is a universal reaction to implanted biomaterial that can affect the function and longevity of the implant. A few studies have attempted to identify targets for treating FBR through the use of single-cell RNA sequencing (scRNA-seq), though the generalizability of these findings from an individual study may be limited. In our study, we perform a meta-analysis of scRNA-seq data from all available FBR mouse studies and integrate these data to identify gene signatures specific to FBR across different models and anatomic locations. We identify subclusters of fibroblasts and macrophages that emerge in response to foreign bodies and characterize their signaling pathways, gene ontology terms, and downstream mediators. The fibroblast subpopulations enriched in the setting of FBR demonstrated significant signaling interactions in the transforming growth factor-beta (TGF-β) signaling pathway, with known pro-fibrotic mediators identified as top expressed genes in these FBR-derived fibroblasts. In contrast, FBR-enriched macrophage subclusters highly expressed pro-fibrotic and pro-inflammatory mediators downstream of tumor necrosis factor (TNF) signaling. Cell-cell interactions were additionally interrogated using CellChat, with identification of key signaling interactions enriched between fibroblasts and macrophages in FBR. By combining multiple FBR datasets, our meta-analysis study identifies common cell-specific gene signatures enriched in foreign body reactions, providing potential therapeutic targets for patients requiring medical implants across a myriad of devices and indications.
Collapse
Affiliation(s)
- Norah E. Liang
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (N.E.L.); (J.B.P.); (J.M.L.); (M.J.); (D.C.W.); (M.G.)
- Division of General Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jennifer B. Parker
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (N.E.L.); (J.B.P.); (J.M.L.); (M.J.); (D.C.W.); (M.G.)
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John M. Lu
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (N.E.L.); (J.B.P.); (J.M.L.); (M.J.); (D.C.W.); (M.G.)
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Januszyk
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (N.E.L.); (J.B.P.); (J.M.L.); (M.J.); (D.C.W.); (M.G.)
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Derrick C. Wan
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (N.E.L.); (J.B.P.); (J.M.L.); (M.J.); (D.C.W.); (M.G.)
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michelle Griffin
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (N.E.L.); (J.B.P.); (J.M.L.); (M.J.); (D.C.W.); (M.G.)
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael T. Longaker
- Hagey Laboratory of Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (N.E.L.); (J.B.P.); (J.M.L.); (M.J.); (D.C.W.); (M.G.)
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
39
|
Wang X, Guillem-Marti J, Kumar S, Lee DS, Cabrerizo-Aguado D, Werther R, Alamo KAE, Zhao YT, Nguyen A, Kopyeva I, Huang B, Li J, Hao Y, Li X, Brizuela-Velasco A, Murray A, Gerben S, Roy A, DeForest CA, Springer T, Ruohola-Baker H, Cooper JA, Campbell MG, Manero JM, Ginebra MP, Baker D. De Novo Design of Integrin α5β1 Modulating Proteins for Regenerative Medicine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600123. [PMID: 38979380 PMCID: PMC11230231 DOI: 10.1101/2024.06.21.600123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Integrin α5β1 is crucial for cell attachment and migration in development and tissue regeneration, and α5β1 binding proteins could have considerable utility in regenerative medicine and next-generation therapeutics. We use computational protein design to create de novo α5β1-specific modulating miniprotein binders, called NeoNectins, that bind to and stabilize the open state of α5β1. When immobilized onto titanium surfaces and throughout 3D hydrogels, the NeoNectins outperform native fibronectin and RGD peptide in enhancing cell attachment and spreading, and NeoNectin-grafted titanium implants outperformed fibronectin and RGD-grafted implants in animal models in promoting tissue integration and bone growth. NeoNectins should be broadly applicable for tissue engineering and biomedicine.
Collapse
Affiliation(s)
- Xinru Wang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jordi Guillem-Marti
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya - BarcelonaTech (UPC), Barcelona, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain
| | - Saurav Kumar
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - David S Lee
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Daniel Cabrerizo-Aguado
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya - BarcelonaTech (UPC), Barcelona, Spain
| | - Rachel Werther
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Yan Ting Zhao
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Adam Nguyen
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Graduate Program in Biological Physics, Structure and Design, University of Washington, Seattle, WA, USA
| | - Irina Kopyeva
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Buwei Huang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jing Li
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Yuxin Hao
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Xinting Li
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Aritza Brizuela-Velasco
- DENS-ia Research Group, Faculty of Health Sciences, Miguel de Cervantes European University, Valladolid, Spain
| | - Analisa Murray
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Stacey Gerben
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Anindya Roy
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Cole A DeForest
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
- Department of Chemistry, University of Washington, Seattle, WA, USA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA, USA
| | - Timothy Springer
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Hannele Ruohola-Baker
- Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jonathan A Cooper
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Melody G Campbell
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jose Maria Manero
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya - BarcelonaTech (UPC), Barcelona, Spain
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya - BarcelonaTech (UPC), Barcelona, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain
- Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
40
|
Rappold R, Kalogeropoulos K, Auf dem Keller U, Vogel V, Slack E. Salmonella-driven intestinal edema in mice is characterized by tensed fibronectin fibers. FEBS J 2024; 291:3104-3127. [PMID: 38487972 DOI: 10.1111/febs.17120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/04/2023] [Accepted: 03/05/2024] [Indexed: 07/19/2024]
Abstract
Intestinal edema is a common manifestation of numerous gastrointestinal diseases and is characterized by the accumulation of fluid in the interstitial space of the intestinal wall. Technical advances in laser capture microdissection and low-biomass proteomics now allow us to specifically characterize the intestinal edema proteome. Using advanced proteomics, we identify peptides derived from antimicrobial factors with high signal intensity, but also highlight major contributions from the blood clotting system, extracellular matrix (ECM) and protease-protease inhibitor networks. The ECM is a complex fibrillar network of macromolecules that provides structural and mechanical support to the intestinal tissue. One abundant component of the ECM observed in Salmonella-driven intestinal edema is the glycoprotein fibronectin, recognized for its structure-function interplay regulated by mechanical forces. Using mechanosensitive staining of fibronectin fibers reveals that they are tensed in the edema, despite the high abundance of proteases able to cleave fibronectin. In contrast, fibronectin fibers increasingly relax in other cecal tissue areas as the infection progresses. Co-staining for fibrin(ogen) indicates the formation of a provisional matrix in the edema, similar to what is observed in response to skin injury, while collagen staining reveals a sparse and disrupted collagen fiber network. These observations plus the absence of low tensional fibronectin fibers and the additional finding of a high number of protease inhibitors in the edema proteome could indicate a critical role of stretched fibronectin fibers in maintaining tissue integrity in the severely inflamed cecum. Understanding these processes may also provide valuable functional diagnostic markers of intestinal disease progression in the future.
Collapse
Affiliation(s)
- Ronja Rappold
- Institute of Translational Medicine, ETH Zurich, Switzerland
- Institute of Food, Nutrition and Health, ETH Zurich, Switzerland
| | | | - Ulrich Auf dem Keller
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Viola Vogel
- Institute of Translational Medicine, ETH Zurich, Switzerland
- Botnar Research Center for Child Health, Basel, Switzerland
| | - Emma Slack
- Institute of Food, Nutrition and Health, ETH Zurich, Switzerland
- Botnar Research Center for Child Health, Basel, Switzerland
| |
Collapse
|
41
|
Hung HS, Shen CC, Wu JT, Yueh CY, Yang MY, Yang YC, Cheng WY. Assessment of the Biocompatibility Ability and Differentiation Capacity of Mesenchymal Stem Cells on Biopolymer/Gold Nanocomposites. Int J Mol Sci 2024; 25:7241. [PMID: 39000351 PMCID: PMC11242884 DOI: 10.3390/ijms25137241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/14/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
This study assessed the biocompatibility of two types of nanogold composites: fibronectin-gold (FN-Au) and collagen-gold (Col-Au). It consisted of three main parts: surface characterization, in vitro biocompatibility assessments, and animal models. To determine the structural and functional differences between the materials used in this study, atomic force microscopy, Fourier-transform infrared spectroscopy, and ultraviolet-visible spectrophotometry were used to investigate their surface topography and functional groups. The F-actin staining, proliferation, migration, reactive oxygen species generation, platelet activation, and monocyte activation of mesenchymal stem cells (MSCs) cultured on the FN-Au and Col-Au nanocomposites were investigated to determine their biological and cellular behaviors. Additionally, animal biocompatibility experiments measured capsule formation and collagen deposition in female Sprague-Dawley rats. The results showed that MSCs responded better on the FN-Au and Col-AU nanocomposites than on the control (tissue culture polystyrene) or pure substances, attributed to their incorporation of an optimal Au concentration (12.2 ppm), which induced significant surface morphological changes, nano topography cues, and better biocompatibility. Moreover, neuronal, endothelial, bone, and adipose tissues demonstrated better differentiation ability on the FN-Au and Col-Au nanocomposites. Nanocomposites have a crucial role in tissue engineering and even vascular grafts. Finally, MSCs were demonstrated to effectively enhance the stability of the endothelial structure, indicating that they can be applied as promising alternatives to clinics in the future.
Collapse
Affiliation(s)
- Huey-Shan Hung
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404328, Taiwan (J.-T.W.)
- Translational Medicine Research, China Medical University Hospital, Taichung 404327, Taiwan
| | - Chiung-Chyi Shen
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (C.-C.S.); (Y.-C.Y.)
| | - Jyun-Ting Wu
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404328, Taiwan (J.-T.W.)
| | - Chun-Yu Yueh
- School of Medicine, China Medical University, Taichung 404333, Taiwan
| | - Meng-Yin Yang
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (C.-C.S.); (Y.-C.Y.)
| | - Yi-Chin Yang
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (C.-C.S.); (Y.-C.Y.)
| | - Wen-Yu Cheng
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (C.-C.S.); (Y.-C.Y.)
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
- Taiwan Department of Physical Therapy, Hung Kuang University, Taichung 433304, Taiwan
| |
Collapse
|
42
|
Revert-Ros F, Ventura I, Prieto-Ruiz JA, Hernández-Andreu JM, Revert F. The Versatility of Collagen in Pharmacology: Targeting Collagen, Targeting with Collagen. Int J Mol Sci 2024; 25:6523. [PMID: 38928229 PMCID: PMC11203716 DOI: 10.3390/ijms25126523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/01/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Collagen, a versatile family of proteins with 28 members and 44 genes, is pivotal in maintaining tissue integrity and function. It plays a crucial role in physiological processes like wound healing, hemostasis, and pathological conditions such as fibrosis and cancer. Collagen is a target in these processes. Direct methods for collagen modulation include enzymatic breakdown and molecular binding approaches. For instance, Clostridium histolyticum collagenase is effective in treating localized fibrosis. Polypeptides like collagen-binding domains offer promising avenues for tumor-specific immunotherapy and drug delivery. Indirect targeting of collagen involves regulating cellular processes essential for its synthesis and maturation, such as translation regulation and microRNA activity. Enzymes involved in collagen modification, such as prolyl-hydroxylases or lysyl-oxidases, are also indirect therapeutic targets. From another perspective, collagen is also a natural source of drugs. Enzymatic degradation of collagen generates bioactive fragments known as matrikines and matricryptins, which exhibit diverse pharmacological activities. Overall, collagen-derived peptides present significant therapeutic potential beyond tissue repair, offering various strategies for treating fibrosis, cancer, and genetic disorders. Continued research into specific collagen targeting and the application of collagen and its derivatives may lead to the development of novel treatments for a range of pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | - Fernando Revert
- Mitochondrial and Molecular Medicine Research Group, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain; (F.R.-R.); (I.V.); (J.A.P.-R.); (J.M.H.-A.)
| |
Collapse
|
43
|
Sun GB, Lu YF, Duan XJ. Investigation into the association of FNDC1 and ADAMTS12 gene expression with plumage coloration in Muscovy ducks. Open Life Sci 2024; 19:20220877. [PMID: 38867923 PMCID: PMC11167702 DOI: 10.1515/biol-2022-0877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/08/2024] [Accepted: 04/29/2024] [Indexed: 06/14/2024] Open
Abstract
To elucidate the molecular genetic mechanisms underpinning feather color in Muscovy ducks. A cohort of 100 Muscovy ducks was meticulously selected for this research. Follicular tissues from ducks exhibiting black and white plumage served as the experimental samples. From these tissues, RNA and proteins were extracted for further analysis. The RNA underwent reverse transcription polymerase chain reaction amplification, followed by validation through western blot assays. The data revealed a significant upregulation in the expression of FN domain-containing protein 1 (FNDC1) and ADAMTS12 genes in Muscovy ducks with white plumage traits as opposed to those with black plumage traits. Specifically, individuals with pure white plumage demonstrated a markedly elevated expression of the FNDC1 gene in comparison to their pure black counterparts. Conversely, expression levels of the ADAMTS12 gene were found to be reduced in ducks with pure black plumage relative to those with pure white plumage. Notably, the expression patterns of FNDC1 and ADAMTS12 genes exhibited inconsistencies between mRNA and protein levels. This study offers significant insights into the molecular genetic mechanisms underlying feather color variation in Muscovy ducks. FNDC1 and ADAMTS12 could be considered potential targets for genetic manipulation or selective breeding strategies aimed at achieving specific feather color phenotypes in Muscovy ducks.
Collapse
Affiliation(s)
- Guo-Bo Sun
- Animal Science and Technology College, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, 225300, Jiangsu, China
| | - Yan-Feng Lu
- Animal Science and Technology College, Jiangsu Agri-animal Husbandry Vocational College, Taizhou, 225300, Jiangsu, China
| | - Xiu-Jun Duan
- Animal Science and Technology College, Jiangsu Agri-animal Husbandry Vocational College, No. 8 of Fenghuang East Road, Hailing District, Taizhou, 225300, Jiangsu, China
| |
Collapse
|
44
|
Roth M, Han B, S’ng CT, Hoang BX, Lambers C. Zinc Iodide Dimethyl Sulfoxide Reduces Collagen Deposition by Increased Matrix Metalloproteinase-2 Expression and Activity in Lung Fibroblasts. Biomedicines 2024; 12:1257. [PMID: 38927463 PMCID: PMC11200730 DOI: 10.3390/biomedicines12061257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Chronic inflammatory lung diseases are characterized by disease-specific extracellular matrix accumulation resulting from an imbalance of matrix metalloproteinases (MMPs) and their inhibitors. Zinc is essential for the function of MMPs, and zinc deficiency has been associated with enhanced tissue remodeling. This study assessed if zinc iodide (ZnI) supplementation through dimethyl sulfoxide (DMSO) modifies the action of MMPs in isolated human lung fibroblasts. The expression and activity of two gelatinases, MMP-2 and MMP-9, were determined by gelatin zymography and enzyme-linked immuno-sorbent assay (ELISA). Collagen degradation was determined by cell-based ELISAs. Collagen type I and fibronectin deposition was stimulated by human recombinant tumor growth factor β1 (TGF-β1). Untreated fibroblasts secreted MMP-2 but only minute amounts of MMP-9. TGF-β1 (5 ng/mL) reduced MMP-2 secretion, but stimulated collagen type I and fibronectin deposition. All the effects of TGF-β1 were significantly reduced in cells treated with ZnI-DMSO over 24 h, while ZnI and DMSO alone had a lower reducing effect. ZnI-DMSO alone did not increase MMP secretion but enhanced the ratio of active to inactive of MMP-2. ZnI alone had a lower enhancing effect than ZnI-DMSO on MMP activity. Furthermore, MMP-2 activity was increased by ZnI-DMSO and ZnI in the absence of cells. Soluble collagen type I increased in the medium of ZnI-DMSO- and ZnI-treated cells. Blocking MMP activity counteracted all the effects of ZnI-DMSO. Conclusion: The data suggest that the combination of ZnI with DMSO reduces fibrotic processes by increasing the degradation of collagen type I by up-regulating the activity of gelatinases. Thus, the combination of ZnI with DMSO might be considered for treatment of fibrotic disorders of the lung. DMSO supported the beneficial effects of ZnI.
Collapse
Affiliation(s)
- Michael Roth
- University Hospital of Basel, University of Basel, 4031 Basel, Switzerland
| | - Bo Han
- Cordoba-Nimni Tissue Engineering and Drug Discovery Lab, Department of Surgery, University of Southern California, Los Angeles, CA 90089, USA
| | | | - Ba Xuan Hoang
- Cordoba-Nimni Tissue Engineering and Drug Discovery Lab, Department of Surgery, University of Southern California, Los Angeles, CA 90089, USA
| | - Christopher Lambers
- Department of Pneumology, Ordensklinikum Linz Elisabethinen, Fadingerstr. 1, 4020 Linz, Austria;
| |
Collapse
|
45
|
Bonadio JD, Bashiri G, Halligan P, Kegel M, Ahmed F, Wang K. Delivery technologies for therapeutic targeting of fibronectin in autoimmunity and fibrosis applications. Adv Drug Deliv Rev 2024; 209:115303. [PMID: 38588958 PMCID: PMC11111362 DOI: 10.1016/j.addr.2024.115303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/29/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Fibronectin (FN) is a critical component of the extracellular matrix (ECM) contributing to various physiological processes, including tissue repair and immune response regulation. FN regulates various cellular functions such as adhesion, proliferation, migration, differentiation, and cytokine release. Alterations in FN expression, deposition, and molecular structure can profoundly impact its interaction with other ECM proteins, growth factors, cells, and associated signaling pathways, thus influencing the progress of diseases such as fibrosis and autoimmune disorders. Therefore, developing therapeutics that directly target FN or its interaction with cells and other ECM components can be an intriguing approach to address autoimmune and fibrosis pathogenesis.
Collapse
Affiliation(s)
- Jacob D Bonadio
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Patrick Halligan
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Michael Kegel
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Fatima Ahmed
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, PA, United States.
| |
Collapse
|
46
|
Sotirova Y, Kiselova-Kaneva Y, Vankova D, Tasinov O, Ivanova D, Popov H, Hristova M, Nikolova K, Andonova V. Tissue Regeneration and Remodeling in Rat Models after Application of Hypericum perforatum L. Extract-Loaded Bigels. Gels 2024; 10:341. [PMID: 38786258 PMCID: PMC11121646 DOI: 10.3390/gels10050341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/12/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
The wound-healing effect of St. John's Wort (SJW) is mainly attributed to hyperforin (HP), but its low stability restricts its topical administration. This study investigates how "free" HP-rich SJW extract (incorporated into a bigel; B/SJW) and extract "protected" by nanostructured lipid carriers (also included in a biphasic semisolid; B/NLC-SJW) affect tissue regeneration in a rat skin excision wound model. Wound diameter, histological changes, and tissue gene expression levels of fibronectin (Fn), matrix metalloproteinase 8 (MMP8), and tumor necrosis factor-alpha (TNF-α) were employed to quantify the healing progress. A significant wound size reduction was achieved after applying both extract-containing semisolids, but after a 21-day application period, the smallest wound size was observed in the B/NLC-SJW-treated animals. However, the inflammatory response was affected more favorably by the bigel containing the "free" SJW extract, as evidenced by histological studies. Moreover, after the application of B/SJW, the expression of Fn, MMP8, and TNF-α was significantly higher than in the positive control. In conclusion, both bigel formulations exhibited beneficial effects on wound healing in rat skin, but B/SJW affected skin restoration processes in a comprehensive and more efficient way.
Collapse
Affiliation(s)
- Yoana Sotirova
- Department of Pharmaceutical Technologies, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria;
| | - Yoana Kiselova-Kaneva
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria (O.T.); (D.I.)
| | - Deyana Vankova
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria (O.T.); (D.I.)
| | - Oskan Tasinov
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria (O.T.); (D.I.)
| | - Diana Ivanova
- Department of Biochemistry, Molecular Medicine and Nutrigenomics, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria (O.T.); (D.I.)
| | - Hristo Popov
- Department of General and Clinical Pathology, Forensic Medicine and Deontology, Faculty of Medicine, Medical University of Varna, 9000 Varna, Bulgaria;
| | - Minka Hristova
- Department of Physiology and Pathophysiology, Faculty of Medicine, Medical University of Varna, 9000 Varna, Bulgaria;
| | - Krastena Nikolova
- Department of Physics and Biophysics, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria;
| | - Velichka Andonova
- Department of Pharmaceutical Technologies, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria;
| |
Collapse
|
47
|
Di T, Feng C, Wang L, Xu J, Du Y, Cheng B, Chen Y, Wu L. Enhancing Vasculogenesis in Dental Pulp Development: DPSCs-ECs Communication via FN1-ITGA5 Signaling. Stem Cell Rev Rep 2024; 20:1060-1077. [PMID: 38418738 PMCID: PMC11087358 DOI: 10.1007/s12015-024-10695-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Dental pulp regeneration therapy is a challenge to achieve early vascularization during treatment. Studying the regulatory mechanisms of vascular formation during human dental pulp development may provide insights for related therapies. In this study, we utilized single-cell sequencing analysis to compare the gene expression of dental pulp stem cells (DPSCs) and vascular endothelial cells (ECs) from developing and mature dental pulps. METHOD Immunohistochemistry, Western blot, and real-time polymerase chain reaction (RT-PCR) were used to detect fibronectin 1 (FN1) expression and molecules, such as PI3K/AKT. Cell proliferation assay, scratch assay, tube formation assay and were used to investigate the effects of DPSCs on the vasculogenetic capability of ECs. Additionally, animal experiments involving mice were conducted. RESULT The results revealed that DPSCs exist around dental pulp vasculature. FN1 expression was significantly higher in DPSCs from young permanent pulps than mature pulps, promoting HUVEC proliferation, migration, and tube formation via ITGA5 and the downstream PI3K/AKT signaling pathway. CONCLUSION Our data indicate that intercellular communication between DPSCs and ECs mediated by FN1-ITGA5 signaling is crucial for vascularizationduring dental pulp development, laying an experimental foundation for future clinical studies.
Collapse
Affiliation(s)
- Tiankai Di
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
- Department of Stomatology, No.969 Hospital, Joint Logistics Support Force of the Chinese People's Liberation Army, Hohhot, Inner Mongolia, 010000, People's Republic of China
| | - Chao Feng
- Center for Computational Biology, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, 100850, People's Republic of China
- Department of Clinical Laboratory, No.969 Hospital, Joint Logistics Support Force of the Chinese People's Liberation Army, Hohhot, Inner Mongolia, 010000, People's Republic of China
| | - Lulu Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Jinlong Xu
- Department of Stomatology, No.969 Hospital, Joint Logistics Support Force of the Chinese People's Liberation Army, Hohhot, Inner Mongolia, 010000, People's Republic of China
| | - Yang Du
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Baixiang Cheng
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Department of General Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Yujiang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China.
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China.
| | - Lian Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China.
| |
Collapse
|
48
|
Liu L, Hong Y, Ma C, Zhang F, Li Q, Li B, He H, Zhu J, Wang H, Chen L. Circular RNA Gtdc1 Protects Against Offspring Osteoarthritis Induced by Prenatal Prednisone Exposure by Regulating SRSF1-Fn1 Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307442. [PMID: 38520084 PMCID: PMC11132075 DOI: 10.1002/advs.202307442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 02/21/2024] [Indexed: 03/25/2024]
Abstract
Chondrodysplasia is closely associated with low birth weight and increased susceptibility to osteoarthritis in adulthood. Prenatal prednisone exposure (PPE) can cause low birth weight; however, its effect on offspring cartilage development remains unexplored. Herein, rats are administered clinical doses of prednisone intragastrically on gestational days (GDs) 0-20 and underwent long-distance running during postnatal weeks (PWs) 24-28. Knee cartilage is assayed for quality and related index changes on GD20, PW12, and PW28. In vitro experiments are performed to elucidate the mechanism. PPE decreased cartilage proliferation and matrix synthesis, causing offspring chondrodysplasia. Following long-distance running, the PPE group exhibited more typical osteoarthritis-like changes. Molecular analysis revealed that PPE caused cartilage circRNomics imbalance in which circGtdc1 decreased most significantly and persisted postnatally. Mechanistically, prednisolone reduced circGtdc1 expression and binding with Srsf1 to promote degradation of Srsf1 via K48-linked polyubiquitination. This further inhibited the formation of EDA/B+Fn1 and activation of PI3K/AKT and TGFβ pathways, reducing chondrocyte proliferation and matrix synthesis. Finally, intra-articular injection of offspring with AAV-circGtdc1 ameliorated PPE-induced chondrodysplasia, but this effect is reversed by Srsf1 knockout. Altogether, this study confirms that PPE causes chondrodysplasia and susceptibility to osteoarthritis by altering the circGtdc1-Srsf1-Fn1 axis; in vivo, overexpression of circGtdc1 can represent an effective intervention target for ameliorating PPE-induced chondrodysplasia.
Collapse
Affiliation(s)
- Liang Liu
- Department of Orthopedic SurgeryJoint Disease Research Center of Wuhan UniversityZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Yuntian Hong
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Chi Ma
- Department of Orthopedic SurgeryJoint Disease Research Center of Wuhan UniversityZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Fan Zhang
- Department of Orthopedic SurgeryJoint Disease Research Center of Wuhan UniversityZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Qingxian Li
- Department of Orthopedic SurgeryJoint Disease Research Center of Wuhan UniversityZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Bin Li
- Department of Orthopedic SurgeryJoint Disease Research Center of Wuhan UniversityZhongnan Hospital of Wuhan UniversityWuhan430071China
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseWuhan430071China
| | - Hangyuan He
- Department of Orthopedic SurgeryJoint Disease Research Center of Wuhan UniversityZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Jiayong Zhu
- Department of Orthopedic SurgeryJoint Disease Research Center of Wuhan UniversityZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Hui Wang
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseWuhan430071China
- Department of PharmacologyWuhan University School of Basic Medical SciencesWuhan430071China
| | - Liaobin Chen
- Department of Orthopedic SurgeryJoint Disease Research Center of Wuhan UniversityZhongnan Hospital of Wuhan UniversityWuhan430071China
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseWuhan430071China
| |
Collapse
|
49
|
Park H, Patil TV, Dutta SD, Lee J, Ganguly K, Randhawa A, Kim H, Lim KT. Extracellular Matrix-Bioinspired Anisotropic Topographical Cues of Electrospun Nanofibers: A Strategy of Wound Healing through Macrophage Polarization. Adv Healthc Mater 2024; 13:e2304114. [PMID: 38295299 DOI: 10.1002/adhm.202304114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/16/2024] [Indexed: 02/02/2024]
Abstract
The skin serves as the body's outermost barrier and is the largest organ, providing protection not only to the body but also to various internal organs. Owing to continuous exposure to various external factors, it is susceptible to damage that can range from simple to severe, including serious types of wounds such as burns or chronic wounds. Macrophages play a crucial role in the entire wound-healing process and contribute significantly to skin regeneration. Initially, M1 macrophages infiltrate to phagocytose bacteria, debris, and dead cells in fresh wounds. As tissue repair is activated, M2 macrophages are promoted, reducing inflammation and facilitating restoration of the dermis and epidermis to regenerate the tissue. This suggests that extracellular matrix (ECM) promotes cell adhesion, proliferation, migrationand macrophage polarization. Among the numerous strategies, electrospinning is a versatile technique for obtaining ECM-mimicking structures with anisotropic and isotropic topologies of micro/nanofibers. Various electrospun biomaterials influence macrophage polarization based on their isotropic or anisotropic topologies. Moreover, these fibers possess a high surface-area-to-volume ratio, promoting the effective exchange of vital nutrients and oxygen, which are crucial for cell viability and tissue regeneration. Micro/nanofibers with diverse physical and chemical properties can be tailored to polarize macrophages toward skin regeneration and wound healing, depending on specific requirements. This review describes the significance of micro/nanostructures for activating macrophages and promoting wound healing.
Collapse
Affiliation(s)
- Hyeonseo Park
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tejal V Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jieun Lee
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Hojin Kim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| |
Collapse
|
50
|
Buruiană A, Gheban BA, Gheban-Roșca IA, Georgiu C, Crișan D, Crișan M. The Tumor Stroma of Squamous Cell Carcinoma: A Complex Environment That Fuels Cancer Progression. Cancers (Basel) 2024; 16:1727. [PMID: 38730679 PMCID: PMC11083853 DOI: 10.3390/cancers16091727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
The tumor microenvironment (TME), a complex assembly of cellular and extracellular matrix (ECM) components, plays a crucial role in driving tumor progression, shaping treatment responses, and influencing metastasis. This narrative review focuses on the cutaneous squamous cell carcinoma (cSCC) tumor stroma, highlighting its key constituents and their dynamic contributions. We examine how significant changes within the cSCC ECM-specifically, alterations in fibronectin, hyaluronic acid, laminins, proteoglycans, and collagens-promote cancer progression, metastasis, and drug resistance. The cellular composition of the cSCC TME is also explored, detailing the intricate interplay of cancer-associated fibroblasts (CAFs), mesenchymal stem cells (MSCs), endothelial cells, pericytes, adipocytes, and various immune cell populations. These diverse players modulate tumor development, angiogenesis, and immune responses. Finally, we emphasize the TME's potential as a therapeutic target. Emerging strategies discussed in this review include harnessing the immune system (adoptive cell transfer, checkpoint blockade), hindering tumor angiogenesis, disrupting CAF activity, and manipulating ECM components. These approaches underscore the vital role that deciphering TME interactions plays in advancing cSCC therapy. Further research illuminating these complex relationships will uncover new avenues for developing more effective treatments for cSCC.
Collapse
Affiliation(s)
- Alexandra Buruiană
- Department of Pathology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.B.); (C.G.); (D.C.)
| | - Bogdan-Alexandru Gheban
- Department of Histology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
- Emergency Clinical County Hospital, 400347 Cluj-Napoca, Romania
| | - Ioana-Andreea Gheban-Roșca
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400129 Cluj-Napoca, Romania;
| | - Carmen Georgiu
- Department of Pathology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.B.); (C.G.); (D.C.)
| | - Doința Crișan
- Department of Pathology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.B.); (C.G.); (D.C.)
| | - Maria Crișan
- Department of Histology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| |
Collapse
|