1
|
Wu C, Yang B, Chu J. Periostin acts as an oncogene to promote laryngeal cancer progression by activating decorin. Histol Histopathol 2025; 40:687-696. [PMID: 39319525 DOI: 10.14670/hh-18-804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Laryngeal carcinoma (LC) is the second most common malignancy of the head and neck worldwide, with increasing incidence every year. However, the mechanism of its development is not completely clear. Periostin (POSTN) has been reported to be involved in various aspects of tumorigenesis. To determine the influence of POSTN on LC tumorigenesis, we first examined the expression of POSTN in tissues from patients with LC through immunohistochemistry, western blot, and qRT-PCR. Besides, we demonstrated that POSTN promoted LC cell migration, invasion, and proliferation in vitro by CCK-8, colony formation, and Transwell assays, and tumor growth in vivo by immunohistochemistry. Furthermore, the interaction between POSTN and decorin (DCN) was further verified by bioinformatics analysis and immunoprecipitation (IP), finding that POSTN promoted the malignant progression of LC by targeting DCN. Our findings support the idea that the level of POSTN expression and accumulation in tumors correlated with the malignancy degree of LC, suggesting that POSTN may play a potential role in improving laryngeal cancer treatment strategies.
Collapse
Affiliation(s)
- Chao Wu
- Department of Otolaryngology Head and Neck Surgery, Taizhou People's Hospital, Taizhou, Jiangsu Province, China.
| | - Bo Yang
- Department of Otolaryngology Head and Neck Surgery, Taizhou People's Hospital, Taizhou, Jiangsu Province, China
| | - Jiusheng Chu
- Department of Otolaryngology Head and Neck Surgery, Taizhou People's Hospital, Taizhou, Jiangsu Province, China
| |
Collapse
|
2
|
Brothwell JA, Wei Y, Wang J, Guo T, Zhang C, Fortney KR, Duplantier R, Chen L, Batteiger TA, Kaplan MH, Spinola SM, Cao S. A high-resolution view of the immune and stromal cell response to Haemophilus ducreyi infection in human volunteers. mBio 2025; 16:e0388524. [PMID: 39882906 PMCID: PMC11898715 DOI: 10.1128/mbio.03885-24] [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: 12/12/2024] [Accepted: 01/10/2025] [Indexed: 01/31/2025] Open
Abstract
Haemophilus ducreyi causes the genital ulcer disease chancroid and cutaneous ulcers in children. To study its pathogenesis, we developed a human challenge model in which we infect the skin on the upper arm of human volunteers with H. ducreyi to the pustular stage of disease. The model has been used to define lesional architecture, describe the immune infiltrate into the infected sites using flow cytometry, and explore the molecular basis of the immune response using bulk RNA-seq. Here, we used single cell RNA-seq (scRNA-seq) and spatial transcriptomics to simultaneously characterize multiple cell types within infected human skin and determine the cellular origin of differentially expressed transcripts that we had previously identified by bulk RNA-seq. We obtained paired biopsies of pustules and wounded (mock infected) sites from five volunteers for scRNA-seq. We identified 13 major cell types, including T- and NK-like cells, macrophages, dendritic cells, as well as other cell types typically found in the skin. Immune cell types were enriched in pustules, and some subtypes within the major cell types were exclusive to pustules. Sufficient tissue specimens for spatial transcriptomics were available from four of the volunteers. T- and NK-like cells were highly associated with multiple antigen presentation cell types. In pustules, type I interferon stimulation was high in areas that were high in antigen presentation-especially in macrophages near the abscess-compared to wounds. Together, our data provide a high-resolution view of the cellular immune response to the infection of the skin with a human pathogen.IMPORTANCEA high-resolution view of the immune infiltrate due to infection with an extracellular bacterial pathogen in human skin has not yet been defined. Here, we used the human skin pathogen Haemophilus ducreyi in a human challenge model to identify on a single cell level the types of cells that are present in volunteers who fail to spontaneously clear infection and form pustules. We identified 13 major cell types. Immune cells and immune-activated stromal cells were enriched in pustules compared to wounded (mock infected) sites. Pustules formed despite the expression of multiple pro-inflammatory cytokines, such as IL-1β and type I interferon. Interferon stimulation was most evident in macrophages, which were proximal to the abscess. The pro-inflammatory response within the pustule may be tempered by regulatory T cells and cells that express indoleamine 2,3-dioxygenase, leading to failure of the immune system to clear H. ducreyi.
Collapse
Affiliation(s)
- Julie A. Brothwell
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yuhui Wei
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - Jia Wang
- Department of Computer Science, Indiana University, Bloomington, Indiana, USA
| | - Tingbo Guo
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - Chi Zhang
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - Kate R. Fortney
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Rory Duplantier
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Li Chen
- Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Teresa A. Batteiger
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Mark H. Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Stanley M. Spinola
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sha Cao
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
3
|
Ding X, Xie S, Zhang W, Zhu Y, Xu D, Xian S, Sun H, Guo X, Li Y, Lu J, Tong X, Huang R, Ji S, Xia Z. Current application of tissue-engineered dermal scaffolds mimicking the extracellular matrix microenvironment in wound healing. Regen Ther 2025; 28:371-382. [PMID: 39896445 PMCID: PMC11786805 DOI: 10.1016/j.reth.2024.12.018] [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: 10/20/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
With the continuous advancement of materials science, cell biology, and biotechnology, tissue engineering has introduced novel solutions to traditional wound healing approaches, particularly demonstrating significant potential in addressing complex or non-healing wounds. One of the key technologies in this field, dermal scaffolds, serve as wound coverage materials that mimic the structural framework of the dermis. They primarily assume the function of extracellular matrix, providing space for cell attachment, migration, and proliferation, thus supporting cellular growth and regulating multiple biological processes in healing. Tissue engineering utilizes combinations of natural or synthetic scaffolds, seeded cells, or growth factors to induce distinct effects in angiogenesis, extracellular matrix deposition, and functional recovery. Therefore, various bioengineered dermal scaffolds hold significant potential for clinical translation in wound healing. This review outlines various extracellular matrix molecules utilized in the development of dermal scaffolds, emphasizes recent progress in cell- and growth factor-modified scaffolds, and discusses the challenges and future perspectives in this evolving field.
Collapse
Affiliation(s)
| | | | | | - Yushu Zhu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Dayuan Xu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Shuyuan Xian
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Hanlin Sun
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Xinya Guo
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Yixu Li
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Jianyu Lu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Xirui Tong
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| | - Zhaofan Xia
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, No. 168 Changhai Road, Shanghai 200433, China
| |
Collapse
|
4
|
Ross GR, Vodanovic-Jankovic S, Benjamin IJ. Geranylgeranyl Pyrophosphate Promotes Profibrotic Factors and Collagen-Specific Chaperone HSP47 in Fibroblasts. J Cell Mol Med 2024; 28:e70273. [PMID: 39716037 DOI: 10.1111/jcmm.70273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 12/25/2024] Open
Abstract
Fibrosis, characterised by excessive extracellular matrix deposition, contributes to both organ failure and significant mortality worldwide. Whereas fibroblasts are activated into myofibroblasts, marked by phenotypic factors such as α-smooth muscle actin (α-SMA), periostin, fibroblast activation protein (FAP) and heat shock protein 47 (HSP47), the cellular processes of trans-differentiation for fibrosis development remain poorly understood. Herein, we hypothesised that the molecular signalling of geranylgeranyl pyrophosphate (GGPP), a crucial biochemical molecule for protein prenylation, is essential in the regulation of profibrotic mechanisms for fibroblast-to-myofibroblast activation. To test this hypothesis, we demonstrated pharmacological inhibition of geranylgeranyl pyrophosphate synthase (GGPS1) significantly decreased TGF-β1-dependent myofibroblast differentiation assessed by reduced α-SMA, periostin, FAP and HSP47 expression. Exogenous GGPP in the presence of GGPS1 inhibition restored TGF-β1-induced differentiation, supporting posttranslational requirements of GGPP modification during myofibroblast differentiation. Selective inhibition of either geranylgeranyl transferase or farnesyl transferase significantly impacted TGF-β1-induced myofibroblast α-SMA and HSP47 expression. The importance of protein prenylation as a key regulator of myofibroblast differentiation was remarkably revealed by an unexpected decrease in HSP47 expression. In contrast, direct HSP47 inhibition not only suppressed TGF-β1-induced α-SMA expression but surprisingly could not be rescued using exogenous GGPP. A selective role for the ER-resident chaperone HSP47 expression downstream of GGPP was suggested when the effects of GGPS1 inhibition on periostin expression were counteracted by GGPP and geranylgeranyl transferase inhibition. Taken together, our findings underscore for the first time the functional role of cholesterol synthesis-independent GGPP-dependent pathway in fibroblast-to-myofibroblast transition and open new potential therapeutic targets for antifibrosis therapies.
Collapse
Affiliation(s)
- Gracious R Ross
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | - Ivor J Benjamin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
5
|
Tian J, Liu X, Zhu D, Li J. Periostin regulates the activity of keloid fibroblasts by activating the JAK/STAT signaling pathway. Heliyon 2024; 10:e38821. [PMID: 39524886 PMCID: PMC11543875 DOI: 10.1016/j.heliyon.2024.e38821] [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: 03/05/2024] [Revised: 09/05/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
A keloid is secondary to skin trauma or has spontaneously manifested as an overgrowth and occurs when the skin heals abnormally after an injury. The main pathological manifestations are abnormal proliferation of keloid fibroblasts (KEL-FIB). This study researched periostin (POSTN) on keloid fibroblasts (KEL-FIB) and the associated mechanism, aiming to provide a reference for the targeted therapy of keloid. We got tissues from Second People's Hospital of Guangxi Zhuang Autonomous Region between June 2022 and March 2023. POSTN expression was increased in keloid skin tissue and KEL-FIB than normal skin tissue and normal fibroblasts. We collected and inoculated KEL-FIB cells, transfection of si-NC (Silencing of POSTN negative control), si-POSTN (Silencing of POSTN), pcDNA-NC (Overexpression of POSTN negative control), and POSTN (Overexpression of POSTN) (Thermo Fisher Scientific) used Lipofectamine 2000 transfection reagent. Wound closure, cell proliferation viability, migrated cell numbers, and POSTN, p-JAK2, p-STAT3 protein levels were reduced in the si-POSTN group. Wound closure, cell proliferation viability, migrated cell numbers, and POSTN, p-JAK2, p-STAT3 protein levels were elevated in the POSTN group. POSTN protein levels did not changed and wound closure, cell proliferation viability, migrated cell numbers, were reduced in the POSTN + S-Ruxolitinib group. The study results indicated that POSTN promotes cell migration and proliferation by activating the JAK/STAT pathway, promoting KEL-FIB development.
Collapse
Affiliation(s)
- Jiao Tian
- Zunyi Medical and Pharmaceutical College, Zunyi City, 563006, China
| | - Xin Liu
- Department of Burn Plastic Surgery, Zunyi Medical University Hospital, Zunyi City, 563000, China
| | - Dawei Zhu
- Zunyi Medical and Pharmaceutical College, Zunyi City, 563006, China
| | - Jianyi Li
- Department of Burn Plastic Surgery and Wound Repair, second People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi City, 541002, China
| |
Collapse
|
6
|
McLeod KA, Di Gregorio M, Tinney D, Carmichael J, Zuanazzi D, Siqueira WL, Rizkalla A, Hamilton DW. Galectin-3/Gelatin Electrospun Scaffolds Modulate Collagen Synthesis in Skin Healing but Do Not Improve Wound Closure Kinetics. Bioengineering (Basel) 2024; 11:960. [PMID: 39451336 PMCID: PMC11504234 DOI: 10.3390/bioengineering11100960] [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: 08/16/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Chronic wounds remain trapped in a pro-inflammatory state, with strategies targeted at inducing re-epithelialization and the proliferative phase of healing desirable. As a member of the lectin family, galectin-3 is implicated in the regulation of macrophage phenotype and epithelial migration. We investigated if local delivery of galectin-3 enhanced skin healing in a full-thickness excisional C57BL/6 mouse model. An electrospun gelatin scaffold loaded with galectin-3 was developed and compared to topical delivery of galectin-3. Electrospun gelatin/galectin-3 scaffolds had an average fiber diameter of 200 nm, with 83% scaffold porosity approximately and an average pore diameter of 1.15 μm. The developed scaffolds supported dermal fibroblast adhesion, matrix deposition, and proliferation in vitro. In vivo treatment of 6 mm full-thickness excisional wounds with gelatin/galectin-3 scaffolds did not influence wound closure, re-epithelialization, or macrophage phenotypes, but increased collagen synthesis. In comparison, topical delivery of galectin-3 [6.7 µg/mL] significantly increased arginase-I cell density at day 7 versus untreated and gelatin/galectin-3 scaffolds (p < 0.05). A preliminary assessment of increasing the concentration of topical galectin-3 demonstrated that at day 7, galectin-3 [12.5 µg/mL] significantly increased both epithelial migration and collagen content in a concentration-dependent manner. In conclusion, local delivery of galectin 3 shows potential efficacy in modulating skin healing in a concentration-dependent manner.
Collapse
Affiliation(s)
- Karrington A. McLeod
- Faculty of Engineering, School of Biomedical Engineering, University of Western Ontario, London, ON N6A 3K7, Canada (A.R.)
| | - Madeleine Di Gregorio
- Faculty of Engineering, School of Biomedical Engineering, University of Western Ontario, London, ON N6A 3K7, Canada (A.R.)
| | - Dylan Tinney
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada; (D.T.); (J.C.)
| | - Justin Carmichael
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada; (D.T.); (J.C.)
| | - David Zuanazzi
- Biochemistry Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (D.Z.); (W.L.S.)
| | - Walter L. Siqueira
- Biochemistry Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (D.Z.); (W.L.S.)
- College of Dentistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Amin Rizkalla
- Faculty of Engineering, School of Biomedical Engineering, University of Western Ontario, London, ON N6A 3K7, Canada (A.R.)
- School of Dentistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Douglas W. Hamilton
- Faculty of Engineering, School of Biomedical Engineering, University of Western Ontario, London, ON N6A 3K7, Canada (A.R.)
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada; (D.T.); (J.C.)
- School of Dentistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
7
|
Rusbjerg-Weberskov CE, Gant MS, Chamot-Rooke J, Nielsen NS, Enghild JJ. Development of a top-down MS assay for specific identification of human periostin isoforms. Front Mol Biosci 2024; 11:1399225. [PMID: 38962283 PMCID: PMC11220192 DOI: 10.3389/fmolb.2024.1399225] [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: 03/11/2024] [Accepted: 05/31/2024] [Indexed: 07/05/2024] Open
Abstract
Periostin is a matricellular protein encoded by the POSTN gene that is alternatively spliced to produce ten different periostin isoforms with molecular weights ranging from 78 to 91 kDa. It is known to promote fibrillogenesis, organize the extracellular matrix, and bind integrin-receptors to induce cell signaling. As well as being a key component of the wound healing process, it is also known to participate in the pathogenesis of different diseases including atopic dermatitis, asthma, and cancer. In both health and disease, the functions of the different periostin isoforms are largely unknown. The ability to precisely determine the isoform profile of a given human sample is fundamental for characterizing their functional significance. Identification of periostin isoforms is most often carried out at the transcriptional level using RT-PCR based approaches, but due to high sequence homogeneity, identification on the protein level has always been challenging. Top-down proteomics, where whole proteins are measured by mass spectrometry, offers a fast and reliable method for isoform identification. Here we present a fully developed top-down mass spectrometry assay for the characterization of periostin splice isoforms at the protein level.
Collapse
Affiliation(s)
| | - Megan S. Gant
- Mass Spectrometry for Biology, Institut Pasteur, Université Paris Cité, CNRS UAR 2024, Paris, France
| | - Julia Chamot-Rooke
- Mass Spectrometry for Biology, Institut Pasteur, Université Paris Cité, CNRS UAR 2024, Paris, France
| | - Nadia Sukusu Nielsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan J. Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
8
|
Ren Y, Wan R, Zhao G, Kuroiwa T, Moran SL, Gingery A, Zhao C. Gene expression of Postn and FGF7 in canine chordae tendineae and their effects on flexor tenocyte biology. J Orthop Res 2024; 42:961-972. [PMID: 37990927 DOI: 10.1002/jor.25745] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 11/23/2023]
Abstract
Chordae tendineae, referred to as heart tendinous cords, act as tendons connecting the papillary muscles to the valves in the heart. Their role is analogous to tendons in the musculoskeletal system. Despite being exposed to millions of cyclic tensile stretches over a human's lifetime, chordae tendineae rarely suffer from overuse injuries. On the other hand, musculoskeletal tendinopathy is very common and remains challenging in clinical treatment. The objective of this study was to investigate the mechanism behind the remarkable durability and resistance to overuse injuries of chordae tendineae, as well as to explore their effects on flexor tenocyte biology. The messenger RNA expression profiles of chordae tendineae were analyzed using RNA sequencing and verified by quantitative reverse transcription polymerase chain reaction and immunohistochemistry. Interestingly, we found that periostin (Postn) and fibroblast growth factor 7 (FGF7) were expressed at significantly higher levels in chordae tendineae, compared to flexor tendons. We further treated flexor tenocytes in vitro with periostin and FGF7 to examine their effects on the proliferation, migration, apoptosis, and tendon-related gene expression of flexor tenocytes. The results displayed enhanced cell proliferation ability at an early stage and an antiapoptotic effect on tenocytes, while treated with periostin and/or FGF7 proteins. Furthermore, there was a trend of promoted tenocyte migration capability. These findings indicated that Postn and FGF7 may represent novel cytokines to target flexor tendon healing. Clinical significance: The preliminary discovery leads to a novel idea for treating tendinopathy in the musculoskeletal system using specific molecules identified from chordae tendineae.
Collapse
Affiliation(s)
- Ye Ren
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Rou Wan
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Gongyin Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Department of Orthopedic Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Tomoyuki Kuroiwa
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Steven L Moran
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Anne Gingery
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
9
|
Jones DC, Jollands MC, D'Haenens-Johansson UFS, Muchnikov AB, Tsai TH. Development of a large volume line scanning, high spectral range and resolution 3D hyperspectral photoluminescence imaging microscope for diamond and other high refractive index materials. OPTICS EXPRESS 2024; 32:15231-15242. [PMID: 38859179 DOI: 10.1364/oe.516046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/26/2024] [Indexed: 06/12/2024]
Abstract
Hyperspectral photoluminescence (PL) imaging is a powerful technique that can be used to understand the spatial distribution of emitting species in many materials. Volumetric hyperspectral imaging of weakly emitting color centers often necessitates considerable data collection times when using commercial systems. We report the development of a line-scanning hyperspectral imaging microscope capable of measuring the luminescence emission spectra for diamond volumes up to 2.20 × 30.00 × 6.30 mm with a high lateral spatial resolution of 1-3 µm. In an single X-λ measurement, spectra covering a 711 nm range, in a band from 400-1100 nm, with a spectral resolution up to 0.25 nm can be acquired. Data sets can be acquired with 723 (X) × 643 (Y) × 1172 (λ) pixels at a rate of 6 minutes/planar image slice, allowing for volumetric hyperspectral imaging with high sampling. This instrument demonstrates the ability to detect emission from several different color centers in diamond both at the surface and internally, providing a non-destructive method to probe their 3D spatial distribution, and is currently not achievable with any other commonly used system or technique.
Collapse
|
10
|
Yujra VQ, Silveira EJDD, Ribeiro DA, Castilho RM, Squarize CH. Clock gene Per2 modulates epidermal tissue repair in vivo. J Cell Biochem 2024; 125:e30513. [PMID: 38229522 PMCID: PMC10932909 DOI: 10.1002/jcb.30513] [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: 08/28/2023] [Revised: 11/18/2023] [Accepted: 12/05/2023] [Indexed: 01/18/2024]
Abstract
Wound healing can be influenced by genes that control the circadian cycle, including Per2 and BMAL1, which coordinate the functions of several organs, including the skin. The aim of the study was to evaluate the role of PER2 during experimental skin wound healing. Two groups (control and Per2-KO), consisting of 14 male mice each, were anesthetized by inhalation, and two 6 mm wounds were created on their dorsal skin using a punch biopsy. A silicone ring was sutured around the wound perimeter to restrict contraction. The wound healing process was clinically measured daily (closure index) until complete wound repair. On Day 6, histomorphometric analysis was performed using the length and thickness of the epithelial migration tongue, in addition to counting vessels underlying the lesion by immunofluorescence assay and maturation of collagen fibers through picrosirius staining. Bromodeoxyuridine (BrdU) incorporation and quantification were performed using the subcutaneous injection technique 2 h before euthanasia and through immunohistochemical analysis of the proliferative index. In addition, the qualitative analysis of myofibroblasts and periostin distribution in connective tissue was performed by immunofluorescence. Statistically significant differences were observed in the healing time between the experimental groups (means: 15.5 days for control mice and 13.5 days for Per2-KO; p = 0.001). The accelerated healing observed in the Per2-KO group (p < 0.05) was accompanied by statistical differences in wound diameter and length of the migrating epithelial tongue (p = 0.01) compared to the control group. Regarding BrdU immunoreactivity, higher expression was observed in the intact epithelium of Per2-KO animals (p = 0.01), and this difference compared to control was also present, to a lesser extent, at the wound site (p = 0.03). Immunofluorescence in the connective tissue underlying the wound showed a higher angiogenic potential in the Per2-KO group in the intact tissue area and the wound region (p < 0.01), where increased expression of myofibroblasts was also observed. Qualitative analysis revealed the distribution of periostin protein and collagen fibers in the connective tissue underlying the wound, with greater organization and maturation during the analyzed period. Our research showed that the absence of the Per2 gene positively impacts the healing time of the skin in vivo. This acceleration depends on the increase of epithelial proliferative and angiogenic capacity of cells carrying the Per2 deletion.
Collapse
Affiliation(s)
- Veronica Quispe Yujra
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan (UM), Ann Arbor, Michigan, USA
- Department of Biosciences, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Ericka Janine Dantas da Silveira
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan (UM), Ann Arbor, Michigan, USA
- Odontology Sciences Postgraduate Program, Dentistry Department, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil
| | - Daniel Araki Ribeiro
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan (UM), Ann Arbor, Michigan, USA
- Department of Biosciences, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Rogerio Moraes Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan (UM), Ann Arbor, Michigan, USA
| | - Cristiane Helena Squarize
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan (UM), Ann Arbor, Michigan, USA
| |
Collapse
|
11
|
Ackerman JE, Muscat SN, Adjei-Sowah E, Korcari A, Nichols AEC, Buckley MR, Loiselle AE. Identification of Periostin as a critical niche for myofibroblast dynamics and fibrosis during tendon healing. Matrix Biol 2024; 125:59-72. [PMID: 38101460 PMCID: PMC10922883 DOI: 10.1016/j.matbio.2023.12.004] [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: 07/24/2023] [Revised: 11/17/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023]
Abstract
Tendon injuries are a major clinical problem, with poor patient outcomes caused by abundant scar tissue deposition during healing. Myofibroblasts play a critical role in the initial restoration of structural integrity after injury. However, persistent myofibroblast activity drives the transition to fibrotic scar tissue formation. As such, disrupting myofibroblast persistence is a key therapeutic target. While myofibroblasts are typically defined by the presence of αSMA+ stress fibers, αSMA is expressed in other cell types including the vasculature. As such, modulation of myofibroblast dynamics via disruption of αSMA expression is not a translationally tenable approach. Recent work has demonstrated that Periostin-lineage (PostnLin) cells are a precursor for cardiac fibrosis-associated myofibroblasts. In contrast to this, here we show that PostnLin cells contribute to a transient αSMA+ myofibroblast population that is required for functional tendon healing, and that Periostin forms a supportive matrix niche that facilitates myofibroblast differentiation and persistence. Collectively, these data identify the Periostin matrix niche as a critical regulator of myofibroblast fate and persistence that could be targeted for therapeutic manipulation to facilitate regenerative tendon healing.
Collapse
Affiliation(s)
- Jessica E Ackerman
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States; NDORMS, University of Oxford, Oxford, United Kingdom
| | - Samantha N Muscat
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States; Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Emmanuela Adjei-Sowah
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States; Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Antonion Korcari
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States; Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Anne E C Nichols
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States; Department of Orthopaedics & Physical Performance, University of Rochester Medical Center, Rochester, NY, United States
| | - Mark R Buckley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States; Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Alayna E Loiselle
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States; Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States; Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States; Department of Orthopaedics & Physical Performance, University of Rochester Medical Center, Rochester, NY, United States.
| |
Collapse
|
12
|
Abstract
Cardiac fibrosis, which is the buildup of proteins in the connective tissues of the heart, can lead to end-stage extracellular matrix (ECM) remodeling and ultimately heart failure. Cardiac remodeling involves changes in gene expression in cardiac cells and ECM, which significantly leads to the morbidity and mortality in heart failure. However, despite extensive research, the elusive intricacies underlying cardiac fibrosis remain unidentified. Periostin, an extracellular matrix (ECM) protein of the fasciclin superfamily, acts as a scaffold for building complex architectures in the ECM, which improves intermolecular interactions and augments the mechanical properties of connective tissues. Recent research has shown that periostin not only contributes to normal ECM homeostasis in a healthy heart but also serves as a potent inducible regulator of cellular reorganization in cardiac fibrosis. Here, we reviewed the constitutive domain of periostin and its interaction with other ECM proteins. We have also discussed the critical pathophysiological functions of periostin in cardiac remodeling mechanisms, including two distinct yet potentially intertwined mechanisms. Furthermore, we will focus on the intrinsic complexities within periostin research, particularly surrounding the contentious issues observed in experimental findings.
Collapse
Affiliation(s)
- Bao Qiao
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xuehao Liu
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Bailu Wang
- Clinical Trial Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Shujian Wei
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
13
|
Raja E, Clarin MTRDC, Yanagisawa H. Matricellular Proteins in the Homeostasis, Regeneration, and Aging of Skin. Int J Mol Sci 2023; 24:14274. [PMID: 37762584 PMCID: PMC10531864 DOI: 10.3390/ijms241814274] [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: 08/31/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Matricellular proteins are secreted extracellular proteins that bear no primary structural functions but play crucial roles in tissue remodeling during development, homeostasis, and aging. Despite their low expression after birth, matricellular proteins within skin compartments support the structural function of many extracellular matrix proteins, such as collagens. In this review, we summarize the function of matricellular proteins in skin stem cell niches that influence stem cells' fate and self-renewal ability. In the epidermal stem cell niche, fibulin 7 promotes epidermal stem cells' heterogeneity and fitness into old age, and the transforming growth factor-β-induced protein ig-h3 (TGFBI)-enhances epidermal stem cell growth and wound healing. In the hair follicle stem cell niche, matricellular proteins such as periostin, tenascin C, SPARC, fibulin 1, CCN2, and R-Spondin 2 and 3 modulate stem cell activity during the hair cycle and may stabilize arrector pili muscle attachment to the hair follicle during piloerections (goosebumps). In skin wound healing, matricellular proteins are upregulated, and their functions have been examined in various gain-and-loss-of-function studies. However, much remains unknown concerning whether these proteins modulate skin stem cell behavior, plasticity, or cell-cell communications during wound healing and aging, leaving a new avenue for future studies.
Collapse
Affiliation(s)
- Erna Raja
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan; (E.R.); (M.T.R.D.C.C.)
| | - Maria Thea Rane Dela Cruz Clarin
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan; (E.R.); (M.T.R.D.C.C.)
- Ph.D. Program in Humanics, School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba 305-8577, Japan
| | - Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan; (E.R.); (M.T.R.D.C.C.)
| |
Collapse
|
14
|
Ackerman JE, Adjei-Sowah E, Korcari A, Muscat SN, Nichols AE, Buckley MR, Loiselle AE. Identification of Periostin as a critical niche for myofibroblast dynamics and fibrosis during tendon healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.21.550090. [PMID: 37502924 PMCID: PMC10370208 DOI: 10.1101/2023.07.21.550090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Tendon injuries are a major clinical problem, with poor patient outcomes caused by abundant scar tissue deposition during healing. Myofibroblasts play a critical role in the initial restoration of structural integrity after injury. However, persistent myofibroblast activity drives the transition to fibrotic scar tissue formation. As such, disrupting myofibroblast persistence is a key therapeutic target. While myofibroblasts are typically defined by the presence of αSMA+ stress fibers, αSMA is expressed in other cell types including the vasculature. As such, modulation of myofibroblast dynamics via disruption of αSMA expression is not a translationally tenable approach. Recent work has demonstrated that Periostin-lineage (PostnLin) cells are a precursor for cardiac fibrosis-associated myofibroblasts. In contrast to this, here we show that PostnLin cells contribute to a transient αSMA+ myofibroblast population that is required for functional tendon healing, and that Periostin forms a supportive matrix niche that facilitates myofibroblast differentiation and persistence. Collectively, these data identify the Periostin matrix niche as a critical regulator of myofibroblast fate and persistence that could be targeted for therapeutic manipulation to facilitate regenerative tendon healing.
Collapse
Affiliation(s)
- Jessica E. Ackerman
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY
- Current affiliation: NDORMS, University of Oxford, Oxford, United Kingdom
| | - Emmanuela Adjei-Sowah
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY
- Department of Biomedical Engineering, University of Rochester, Rochester, NY
| | - Antonion Korcari
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY
- Department of Biomedical Engineering, University of Rochester, Rochester, NY
| | - Samantha N. Muscat
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY
| | - Anne E.C. Nichols
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY
- Department of Orthopaedics & Physical Performance, University of Rochester Medical Center, Rochester, NY
| | - Mark R. Buckley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY
- Department of Biomedical Engineering, University of Rochester, Rochester, NY
| | - Alayna E. Loiselle
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY
- Department of Biomedical Engineering, University of Rochester, Rochester, NY
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY
- Department of Orthopaedics & Physical Performance, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
15
|
Nagalingam RS, Chattopadhyaya S, Al-Hattab DS, Cheung DYC, Schwartz LY, Jana S, Aroutiounova N, Ledingham DA, Moffatt TL, Landry NM, Bagchi RA, Dixon IMC, Wigle JT, Oudit GY, Kassiri Z, Jassal DS, Czubryt MP. Scleraxis and fibrosis in the pressure-overloaded heart. Eur Heart J 2022; 43:4739-4750. [PMID: 36200607 DOI: 10.1093/eurheartj/ehac362] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 06/02/2022] [Accepted: 06/23/2022] [Indexed: 01/05/2023] Open
Abstract
AIMS In response to pro-fibrotic signals, scleraxis regulates cardiac fibroblast activation in vitro via transcriptional control of key fibrosis genes such as collagen and fibronectin; however, its role in vivo is unknown. The present study assessed the impact of scleraxis loss on fibroblast activation, cardiac fibrosis, and dysfunction in pressure overload-induced heart failure. METHODS AND RESULTS Scleraxis expression was upregulated in the hearts of non-ischemic dilated cardiomyopathy patients, and in mice subjected to pressure overload by transverse aortic constriction (TAC). Tamoxifen-inducible fibroblast-specific scleraxis knockout (Scx-fKO) completely attenuated cardiac fibrosis, and significantly improved cardiac systolic function and ventricular remodelling, following TAC compared to Scx+/+ TAC mice, concomitant with attenuation of fibroblast activation. Scleraxis deletion, after the establishment of cardiac fibrosis, attenuated the further functional decline observed in Scx+/+ mice, with a reduction in cardiac myofibroblasts. Notably, scleraxis knockout reduced pressure overload-induced mortality from 33% to zero, without affecting the degree of cardiac hypertrophy. Scleraxis directly regulated transcription of the myofibroblast marker periostin, and cardiac fibroblasts lacking scleraxis failed to upregulate periostin synthesis and secretion in response to pro-fibrotic transforming growth factor β. CONCLUSION Scleraxis governs fibroblast activation in pressure overload-induced heart failure, and scleraxis knockout attenuated fibrosis and improved cardiac function and survival. These findings identify scleraxis as a viable target for the development of novel anti-fibrotic treatments.
Collapse
Affiliation(s)
- Raghu S Nagalingam
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - Sikta Chattopadhyaya
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - Danah S Al-Hattab
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - David Y C Cheung
- Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - Leah Y Schwartz
- Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - Sayantan Jana
- Department of Physiology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Nina Aroutiounova
- Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - D Allison Ledingham
- Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - Teri L Moffatt
- Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - Natalie M Landry
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - Rushita A Bagchi
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Ian M C Dixon
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| | - Jeffrey T Wigle
- Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada.,Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Gavin Y Oudit
- Department of Physiology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada.,Division of Cardiology, Department of Medicine, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Davinder S Jassal
- Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada.,Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Michael P Czubryt
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, Canada
| |
Collapse
|
16
|
Cárdenas-León CG, Mäemets-Allas K, Klaas M, Lagus H, Kankuri E, Jaks V. Matricellular proteins in cutaneous wound healing. Front Cell Dev Biol 2022; 10:1073320. [PMID: 36506087 PMCID: PMC9730256 DOI: 10.3389/fcell.2022.1073320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022] Open
Abstract
Cutaneous wound healing is a complex process that encompasses alterations in all aspects of the skin including the extracellular matrix (ECM). ECM consist of large structural proteins such as collagens and elastin as well as smaller proteins with mainly regulative properties called matricellular proteins. Matricellular proteins bind to structural proteins and their functions include but are not limited to interaction with cell surface receptors, cytokines, or protease and evoking a cellular response. The signaling initiated by matricellular proteins modulates differentiation and proliferation of cells having an impact on the tissue regeneration. In this review we give an overview of the matricellular proteins that have been found to be involved in cutaneous wound healing and summarize the information known to date about their functions in this process.
Collapse
Affiliation(s)
| | - Kristina Mäemets-Allas
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Mariliis Klaas
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Heli Lagus
- Department of Plastic Surgery and Wound Healing Centre, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Viljar Jaks
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia,Dermatology Clinic, Tartu University Clinics, Tartu, Estonia,*Correspondence: Viljar Jaks,
| |
Collapse
|
17
|
El-Adili F, Lui JK, Najem M, Farina G, Trojanowska M, Sam F, Bujor AM. Periostin overexpression in scleroderma cardiac tissue and its utility as a marker for disease complications. Arthritis Res Ther 2022; 24:251. [PMID: 36369212 PMCID: PMC9650849 DOI: 10.1186/s13075-022-02943-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 11/02/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To evaluate the levels of periostin in patients with systemic sclerosis (SSc) and their association with features of systemic sclerosis. METHODS The levels of periostin were assessed in the serum of 106 SSc patients and 22 healthy controls and by immunofluorescence staining in cardiac tissue from 4 SSc patients and 4 controls. Serum periostin was measured via enzyme-linked immunosorbent assay. The results were analyzed using Mann-Whitney test or Kruskal-Wallis test followed by Dunn's multiple comparisons tests and Spearman's test for correlations. Cardiac tissue from SSc patients and controls was stained for periostin and co-stained for periostin and collagen type I using immunofluorescence. RESULTS Periostin levels were higher in patients with SSc compared to controls and directly correlated to modified Rodnan skin score and echocardiography parameters of left ventricular measurements. Immunofluorescence staining in SSc cardiac tissue showed patchy periostin expression in all SSc patients, but not in controls. Furthermore, there was extensive periostin expression even in areas without collagen deposition, while all established fibrotic areas showed colocalization of collagen and periostin. There was no association between periostin levels and interstitial lung disease, pulmonary hypertension or other vascular complications. CONCLUSION Periostin is elevated in SSc cardiac tissue in vivo and circulating levels of periostin are increased in SSc, correlating with the extent of disease duration, degree of skin fibrosis, and left ventricular structural assessments. Periostin may be a potential biomarker that can provide further pathogenic insight into cardiac fibrosis in SSc.
Collapse
Affiliation(s)
- Fatima El-Adili
- Division of Rheumatology, Boston University School of Medicine, Boston, MA, USA
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
- Arthritis and Autoimmune Diseases Center, Boston University, 72 E Concord St, Evans 501, Boston, MA, 02118, USA
| | - Justin K Lui
- The Pulmonary Center, Boston University School of Medicine, Boston, MA, USA
| | - Mortada Najem
- Division of Rheumatology, Boston University School of Medicine, Boston, MA, USA
| | - Giuseppina Farina
- Division of Rheumatology, Boston University School of Medicine, Boston, MA, USA
- Arthritis and Autoimmune Diseases Center, Boston University, 72 E Concord St, Evans 501, Boston, MA, 02118, USA
| | - Maria Trojanowska
- Division of Rheumatology, Boston University School of Medicine, Boston, MA, USA
- Arthritis and Autoimmune Diseases Center, Boston University, 72 E Concord St, Evans 501, Boston, MA, 02118, USA
| | - Flora Sam
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Andreea M Bujor
- Division of Rheumatology, Boston University School of Medicine, Boston, MA, USA.
- Arthritis and Autoimmune Diseases Center, Boston University, 72 E Concord St, Evans 501, Boston, MA, 02118, USA.
| |
Collapse
|
18
|
Periostin Modulates Extracellular Matrix Behavior in Tendons. Matrix Biol Plus 2022; 16:100124. [DOI: 10.1016/j.mbplus.2022.100124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 10/10/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
|
19
|
Initial Assessment of Gingival Biotype as a Potential Source of Variability in the Migration, Contraction and Gene Expression of Fibroblasts. Arch Oral Biol 2022; 144:105554. [DOI: 10.1016/j.archoralbio.2022.105554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/22/2022]
|
20
|
Dermal extracellular matrix molecules in skin development, homeostasis, wound regeneration and diseases. Semin Cell Dev Biol 2022; 128:137-144. [PMID: 35339360 DOI: 10.1016/j.semcdb.2022.02.027] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/22/2022] [Indexed: 12/14/2022]
Abstract
The extracellular matrix (ECM) is a dynamic structure that surrounds and anchors cellular components in tissues. In addition to functioning as a structural scaffold for cellular components, ECMs also regulate diverse biological functions, including cell adhesion, proliferation, differentiation, migration, cell-cell interactions, and intracellular signaling events. Dermal fibroblasts (dFBs), the major cellular source of skin ECM, develop from a common embryonic precursor to the highly heterogeneous subpopulations during development and adulthood. Upon injury, dFBs migrate into wound granulation tissue and transdifferentiate into myofibroblasts, which play a critical role in wound contraction and dermal ECM regeneration and deposition. In this review, we describe the plasticity of dFBs during development and wound healing and how various dFB-derived ECM molecules, including collagen, proteoglycans, glycosaminoglycans, fibrillins and matricellular proteins are expressed and regulated, and in turn how these ECM molecules play a role in regulating the function of dFBs and immune cells. Finally, we describe how dysregulation of ECM matrix is associated the pathogenesis of wound healing related skin diseases, including chronic wounds and keloid.
Collapse
|
21
|
The contracture-in-a-well. An in vitro model distinguishes bulk and interfacial processes of irreversible (fibrotic) cell-mediated contraction. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 133:112661. [DOI: 10.1016/j.msec.2022.112661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 11/21/2022]
|
22
|
Wang M, Qian L, Li J, Ming H, Fang L, Li Y, Zhang M, Xu Y, Ban Y, Zhang W, Zhang Y, Liu Y, Wang N. GHSR deficiency exacerbates cardiac fibrosis: role in macrophage inflammasome activation and myofibroblast differentiation. Cardiovasc Res 2021; 116:2091-2102. [PMID: 31790138 DOI: 10.1093/cvr/cvz318] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 08/06/2019] [Accepted: 11/28/2019] [Indexed: 12/19/2022] Open
Abstract
AIMS Sustained activation of β-adrenergic signalling induces cardiac fibrosis, which marks progression to heart failure. GHSR (growth hormone secretagogue receptor) is the receptor for ghrelin, which is an orexigenic gastric hormone with newly defined cardiovascular effects. The present study determined the effects of GHSR deficiency in a mouse model of isoproterenol (ISO)-induced cardiac fibrosis and examined the underlying mechanism. METHODS AND RESULTS Histochemical studies showed that GHSR deficiency exacerbated cardiac fibrosis. Quantitative RT-PCR, western blotting, and immunofluorescence staining demonstrated that cardiac fibroblasts isolated from GHSR-/- mice exhibited increased expression of marker genes for myofibroblast trans-differentiation (α-SMA, SM22, and calponin) upon transforming growth factor-β treatment compared to wild-type mice. RNA-sequencing of heart transcriptomes revealed that differentially expressed genes in GHSR-/- hearts were enriched in such biological processes as extracellular matrix organization, inflammatory response, lipid metabolism, cell cycle, migration, and adhesion. Particularly, GHSR deficiency increased Wnt/β-catenin pathway activation in ISO-induced myocardial fibrosis. In addition, loss of GHSR in macrophages instigated inflammasome activation with increased cleavage and release of interleukin-18. CONCLUSION These results for the first time demonstrated that GHSR deficiency aggravated ISO-induced cardiac fibrosis, suggesting that GHSR was a potential target for the intervention of cardiac fibrosis.
Collapse
Affiliation(s)
- Mo Wang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Lei Qian
- The Advanced Institute of Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Jing Li
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Hao Ming
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Li Fang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Yingjia Li
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Man Zhang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Yaohua Xu
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Yiqian Ban
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Weizhen Zhang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Youyi Zhang
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China.,Institute of Vascular Medicine, The Third Hospital, Peking University, Beijing, China
| | - Yahan Liu
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Nanping Wang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China.,The Advanced Institute of Medical Sciences, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
23
|
Nichols AEC, Muscat SN, Miller SE, Green LJ, Richards MS, Loiselle AE. Impact of isolation method on cellular activation and presence of specific tendon cell subpopulations during in vitro culture. FASEB J 2021; 35:e21733. [PMID: 34160846 DOI: 10.1096/fj.202100405r] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 11/11/2022]
Abstract
Tendon injuries are common and heal poorly, due in part to a lack of understanding of fundamental tendon cell biology. A major impediment to the study of tendon cells is the absence of robust, well-characterized in vitro models. Unlike other tissue systems, current tendon cell models do not account for how differences in isolation methodology may affect the activation state of tendon cells or the presence of various tendon cell subpopulations. The objective of this study was to characterize how common isolation methods affect the behavior, fate, and lineage composition of tendon cell cultures. Tendon cells isolated by explant exhibited reduced proliferative capacity, decreased expression of tendon marker genes, and increased expression of genes associated with fibroblast activation compared to digested cells. Consistently, explanted cells also displayed an increased propensity to differentiate to myofibroblasts compared to digested cells. Explanted cultures from multiple different tendons were substantially enriched for the presence of scleraxis-lineage (Scx-lin+) cells compared to digested cultures, while the overall percentage of S100a4-lineage (S100a4-lin+) cells was dependent on both isolation method and tendon of origin. Neither isolation methods preserved the ratios of Scx-lin+ or S100a4-lin+ to non-lineage cells seen in tendons in vivo. Combined, these data indicate that further refinement of in vitro cultures models is required in order to more accurately understand the effects of various stimuli on tendon cell behavior. Statement of clinical significance: The development of informed in vitro tendon cell models will facilitate enhanced screening of potential therapeutic candidates to improve tendon healing.
Collapse
Affiliation(s)
- Anne E C Nichols
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Samantha N Muscat
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Sarah E Miller
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA
| | - Luke J Green
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA
| | - Michael S Richards
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA
| | - Alayna E Loiselle
- Center for Musculoskeletal Research, Department of Orthopaedics & Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
24
|
Walker JT, Flynn LE, Hamilton DW. Lineage tracing of Foxd1-expressing embryonic progenitors to assess the role of divergent embryonic lineages on adult dermal fibroblast function. FASEB Bioadv 2021; 3:541-557. [PMID: 34258523 PMCID: PMC8255845 DOI: 10.1096/fba.2020-00110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/16/2021] [Accepted: 02/24/2021] [Indexed: 01/10/2023] Open
Abstract
Recent studies have highlighted the functional diversity of dermal fibroblast populations in health and disease, with part of this diversity linked to fibroblast lineage and embryonic origin. Fibroblasts derived from foxd1-expressing progenitors contribute to the myofibroblast populations present in lung and kidney fibrosis in mice but have not been investigated in the context of dermal wound repair. Using a Cre/Lox system to genetically track populations derived from foxd1-expressing progenitors, lineage-positive fibroblasts were identified as a subset of the dermal fibroblast population. During development, lineage-positive cells were most abundant within the dorsal embryonic tissues, contributing to the developing dermal fibroblast population, and remaining in this niche into adulthood. In adult mice, assessment of fibrosis-related gene expression in lineage-positive and lineage-negative populations isolated from wounded and unwounded dorsal skin was performed, identifying an enrichment of transcripts associated with matrix synthesis and remodeling in the lineage-positive populations. Using a novel excisional wound model, ventral skin healed with a greatly reduced frequency of foxd1 lineage-positive cells. This work supports that the embryonic origin of fibroblasts is an important predictor of fibroblast function, but also highlights that within disparate regions, fibroblasts of different lineages likely undergo convergent differentiation contributing to phenotypic similarities.
Collapse
Affiliation(s)
- John T. Walker
- Department of Anatomy and Cell BiologySchulich School of Medicine and DentistryThe University of Western OntarioLondonOntarioCanada
| | - Lauren E. Flynn
- Department of Anatomy and Cell BiologySchulich School of Medicine and DentistryThe University of Western OntarioLondonOntarioCanada
- Department of Chemical and Biochemical EngineeringThompson Engineering BuildingThe University of Western OntarioLondonONCanada
| | - Douglas W. Hamilton
- Department of Anatomy and Cell BiologySchulich School of Medicine and DentistryThe University of Western OntarioLondonOntarioCanada
- Division of Oral BiologySchulich School of Medicine and DentistryThe University of Western OntarioLondonONCanada
| |
Collapse
|
25
|
Nikoloudaki G. Functions of Matricellular Proteins in Dental Tissues and Their Emerging Roles in Orofacial Tissue Development, Maintenance, and Disease. Int J Mol Sci 2021; 22:ijms22126626. [PMID: 34205668 PMCID: PMC8235165 DOI: 10.3390/ijms22126626] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 01/04/2023] Open
Abstract
Matricellular proteins (MCPs) are defined as extracellular matrix (ECM) associated proteins that are important regulators and integrators of microenvironmental signals, contributing to the dynamic nature of ECM signalling. There is a growing understanding of the role of matricellular proteins in cellular processes governing tissue development as well as in disease pathogenesis. In this review, the expression and functions of different MP family members (periostin, CCNs, TSPs, SIBLINGs and others) are presented, specifically in relation to craniofacial development and the maintenance of orofacial tissues, including bone, gingiva, oral mucosa, palate and the dental pulp. As will be discussed, each MP family member has been shown to have non-redundant roles in development, tissue homeostasis, wound healing, pathology and tumorigenesis of orofacial and dental tissues.
Collapse
Affiliation(s)
- Georgia Nikoloudaki
- Schulich Dentistry Department, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; ; Tel.: +1-519-661-2111 (ext. 81102)
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| |
Collapse
|
26
|
Deng CC, Hu YF, Zhu DH, Cheng Q, Gu JJ, Feng QL, Zhang LX, Xu YP, Wang D, Rong Z, Yang B. Single-cell RNA-seq reveals fibroblast heterogeneity and increased mesenchymal fibroblasts in human fibrotic skin diseases. Nat Commun 2021; 12:3709. [PMID: 34140509 PMCID: PMC8211847 DOI: 10.1038/s41467-021-24110-y] [Citation(s) in RCA: 240] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Fibrotic skin disease represents a major global healthcare burden, characterized by fibroblast hyperproliferation and excessive accumulation of extracellular matrix. Fibroblasts are found to be heterogeneous in multiple fibrotic diseases, but fibroblast heterogeneity in fibrotic skin diseases is not well characterized. In this study, we explore fibroblast heterogeneity in keloid, a paradigm of fibrotic skin diseases, by using single-cell RNA-seq. Our results indicate that keloid fibroblasts can be divided into 4 subpopulations: secretory-papillary, secretory-reticular, mesenchymal and pro-inflammatory. Interestingly, the percentage of mesenchymal fibroblast subpopulation is significantly increased in keloid compared to normal scar. Functional studies indicate that mesenchymal fibroblasts are crucial for collagen overexpression in keloid. Increased mesenchymal fibroblast subpopulation is also found in another fibrotic skin disease, scleroderma, suggesting this is a broad mechanism for skin fibrosis. These findings will help us better understand skin fibrotic pathogenesis, and provide potential targets for fibrotic disease therapies.
Collapse
Affiliation(s)
- Cheng-Cheng Deng
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yong-Fei Hu
- Dermatology Hospital, Southern Medical University, Guangzhou, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ding-Heng Zhu
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Qing Cheng
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Jing Gu
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Qing-Lan Feng
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Li-Xue Zhang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Ying-Ping Xu
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Dong Wang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhili Rong
- Dermatology Hospital, Southern Medical University, Guangzhou, China.
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Guangzhou, China.
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
27
|
|
28
|
Su JY, Yu CC, Peng CY, Liao YW, Hsieh PL, Yang LC, Yu CH, Chou MY. Silencing periostin inhibits myofibroblast transdifferentiation of fibrotic buccal mucosal fibroblasts. J Formos Med Assoc 2021; 120:2010-2015. [PMID: 33965260 DOI: 10.1016/j.jfma.2021.04.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/04/2021] [Accepted: 04/08/2021] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND/PURPOSE Oral submucous fibrosis (OSF) a well-recognized oral premalignant disorder. Several studies have demonstrated that periostin, a matricellular protein, is involved in the development and pathogenesis of fibrosis diseases. Nevertheless, the contribution of periostin in OSF remains to be uncovered. The purpose of the study was to illustrate the functional role of periostin involved in OSF pathogenesis. METHODS RNA-sequencing was employed to screen for differentially expressed genes in normal and OSF tissues. Validation of the upregulation of periostin in OSF specimens and fibrotic buccal mucosal fibroblasts (fBMFs) was conducted by qRT-PCR. The correlation of the gene expression of periostin and various fibrosis markers was analyzed. In addition, the functional role of periostin in myofibroblast features was tested using collagen gel contraction and transwell migration assays. RESULTS We observed overexpression of periostin in OSF specimens using RNA-sequencing and confirmed its upregulation in OSF tissues and patient-derived fBMFs. Besides, there was a positive relationship between the expression of periostin and several fibrosis-associated markers, including ACTA2 (α-smooth muscle actin; α-SMA), COL1A1 (type 1 collagen α1 chain), TGFB1 (TGF-β1), and FN1 (fibronectin). Furthermore, we examined the effect of silencing periostin on the maintenance of myofibroblast characteristics and showed that knockdown of periostin suppressed the expression of α-SMA. Also, inhibition of periostin markedly downregulated the myofibroblast activities (collagen gel contraction and migration capacities). CONCLUSION Our results indicate the aberrant expression of periostin in OSF tissues and myofibroblasts. Moreover, the expression of periostin is positively associated with fibrosis markers, and repression of periostin may be a promising direction to alleviate the progression of OSF.
Collapse
Affiliation(s)
- Jyun-Yang Su
- Department of Oral and Maxillofacial Surgery, Changhua Christian Hospital, Changhua, Taiwan; School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Cheng-Chia Yu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan; Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chih-Yu Peng
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Wen Liao
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Pei-Ling Hsieh
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Li-Chiu Yang
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chuan-Hang Yu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan; Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Ming-Yung Chou
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan; Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
29
|
Chaitin H, Lu ML, Wallace MB, Kang Y. Development of a Decellularized Porcine Esophageal Matrix for Potential Applications in Cancer Modeling. Cells 2021; 10:cells10051055. [PMID: 33946915 PMCID: PMC8144998 DOI: 10.3390/cells10051055] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/13/2022] Open
Abstract
Many decellularized extracellular matrix-derived whole organs have been widely used in studies of tissue engineering and cancer models. However, decellularizing porcine esophagus to obtain decellularized esophageal matrix (DEM) for potential biomedical applications has not been widely investigated. In this study a modified decellularization protocol was employed to prepare a porcine esophageal DEM for the study of cancer cell growth. The cellular removal and retention of matrix components in the porcine DEM were fully characterized. The microstructure of the DEM was observed using scanning electronic microscopy. Human esophageal squamous cell carcinoma (ESCC) and human primary esophageal fibroblast cells (FBCs) were seeded in the DEM to observe their growth. Results show that the decellularization process did not cause significant loss of mechanical properties and that blood ducts and lymphatic vessels in the submucosa layer were also preserved. ESCC and FBCs grew on the DEM well and the matrix did not show any toxicity to cells. When FBS and ESCC were cocultured on the matrix, they secreted more periostin, a protein that supports cell adhesion on matrix. This study shows that the modified decellularization protocol can effectively remove the cell materials and maintain the microstructure of the porcine esophageal matrix, which has the potential application of studying cell growth and migration for esophageal cancer models.
Collapse
Affiliation(s)
- Hersh Chaitin
- Department of Biological Science, College of Science, Florida Atlantic University, Boca Raton, FL 33431, USA;
- Department of Biomedical Science, College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA;
| | - Michael L. Lu
- Department of Biomedical Science, College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA;
| | - Michael B. Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Yunqing Kang
- Department of Biomedical Science, College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA;
- Department of Ocean and Mechanical Engineering, College of Computer Science and Engineering, Florida Atlantic University, Boca Raton, FL 33431, USA
- Faculty of Integrative Biology PhD Program, College of Science, Florida Atlantic University, Boca Raton, FL 33431, USA
- Correspondence: ; Tel.: +1-(561)-297-3943
| |
Collapse
|
30
|
Zhang Q, Chang C, Qian C, Xiao W, Zhu H, Guo J, Meng Z, Cui W, Ge Z. Photo-crosslinkable amniotic membrane hydrogel for skin defect healing. Acta Biomater 2021; 125:197-207. [PMID: 33676048 DOI: 10.1016/j.actbio.2021.02.043] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 02/06/2021] [Accepted: 02/25/2021] [Indexed: 01/23/2023]
Abstract
The human amniotic membrane (HAM) collagen matrix derived from human placenta can be decellularized (dHAM) to form a natural biocompatible material. dHAM has different bioactive substances and has been used widely in human tissue engineering research. However, dHAM has some disadvantages, e.g., poor mechanical properties, easy degradation and inconvenient operation and use, so it is not conducive to large-area or full-thickness skin defect healing. To overcome these limitations, for the first time, dHAM was grafted with methacrylic anhydride (MA) to form photocrosslinked dHAM methacrylate (dHAMMA); dHAMMA was then blended with methacrylated gelatin (GelMA), followed by the addition of a photosensitizer for photocrosslinking to obtain the fast-forming GelMA-dHAMMA composite hydrogel. Further, GelMA-dHAMMA was found to have the porous structure of a bicomponent polymer network and good physical and chemical properties. In vitro experiments, GelMA-dHAMMA was found to promote fibroblast proliferation and α-smooth muscle actin (α-SMA) expression. In vivo investigations also demonstrated that GelMA-dHAMMA promotes wound collagen deposition and angiogenesis, and accelerates tissue healing. GelMA-dHAMMA inherits the good mechanical properties of GelMA and maintains the biological activity of the amniotic membrane, promoting the reconstruction and regeneration of skin wounds. Thus, GelMA-dHAMMA can serve as a promising biomaterial in skin tissue engineering. STATEMENT OF SIGNIFICANCE: Since the early 20th century, the human amniotic membrane (HAM) has been successfully used for trauma treatment and reconstruction purposes. dHAM has different bioactive substances and has been used widely in human tissue-engineering research. In this work, the dHAM and gelatin were grafted and modified by using methacrylic anhydride (MA) to form photocrosslinked dHAMMA and methacrylated gelatin (GelMA). Then, the dHAMMA and GelMA were blended with a photosensitizer to form the GelMA-dHAMMA composite hydrogel derived from gelatin-dHAM. GelMA-dHAMMA exhibits a bicomponent-network (BCN) interpenetrating structure. dHAM dydrogel has advantages, e.g., good mechanical properties, slow degradation and convenient operation, so it is conducive to large-area or full-thickness skin defect healing.
Collapse
|
31
|
Musiime M, Chang J, Hansen U, Kadler KE, Zeltz C, Gullberg D. Collagen Assembly at the Cell Surface: Dogmas Revisited. Cells 2021; 10:662. [PMID: 33809734 PMCID: PMC8002325 DOI: 10.3390/cells10030662] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/12/2022] Open
Abstract
With the increased awareness about the importance of the composition, organization, and stiffness of the extracellular matrix (ECM) for tissue homeostasis, there is a renewed need to understand the details of how cells recognize, assemble and remodel the ECM during dynamic tissue reorganization events. Fibronectin (FN) and fibrillar collagens are major proteins in the ECM of interstitial matrices. Whereas FN is abundant in cell culture studies, it is often only transiently expressed in the acute phase of wound healing and tissue regeneration, by contrast fibrillar collagens form a persistent robust scaffold in healing and regenerating tissues. Historically fibrillar collagens in interstitial matrices were seen merely as structural building blocks. Cell anchorage to the collagen matrix was thought to be indirect and occurring via proteins like FN and cell surface-mediated collagen fibrillogenesis was believed to require a FN matrix. The isolation of four collagen-binding integrins have challenged this dogma, and we now know that cells anchor directly to monomeric forms of fibrillar collagens via the α1β1, α2β1, α10β1 and α11β1 integrins. The binding of these integrins to the mature fibrous collagen matrices is more controversial and depends on availability of integrin-binding sites. With increased awareness about the importance of characterizing the total integrin repertoire on cells, including the integrin collagen receptors, the idea of an absolute dependence on FN for cell-mediated collagen fibrillogenesis needs to be re-evaluated. We will summarize data suggesting that collagen-binding integrins in vitro and in vivo are perfectly well suited for nucleating and supporting collagen fibrillogenesis, independent of FN.
Collapse
Affiliation(s)
- Moses Musiime
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway; (M.M.); (C.Z.)
| | - Joan Chang
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (J.C.); (K.E.K.)
| | - Uwe Hansen
- Institute for Musculoskeletal Medicine, University Hospital of Münster, 48149 Münster, Germany;
| | - Karl E. Kadler
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (J.C.); (K.E.K.)
| | - Cédric Zeltz
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway; (M.M.); (C.Z.)
| | - Donald Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway; (M.M.); (C.Z.)
| |
Collapse
|
32
|
Migneault F, Hébert MJ. Autophagy, tissue repair, and fibrosis: a delicate balance. Matrix Biol 2021; 100-101:182-196. [PMID: 33454422 DOI: 10.1016/j.matbio.2021.01.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/12/2022]
Abstract
Tissue repair and fibrosis, an abnormal form of repair, occur in most human organs in response to injury or inflammation. Fibroblasts play a major role in the normal repair process by differentiating into myofibroblasts that synthesize extracellular matrix (ECM) components and favor tissue remodeling to reestablish normal function and integrity. However, their persistent accumulation at the site of injury is a hallmark of fibrosis. Autophagy is a catabolic process that occurs in eukaryotic cells as a stress response to allow cell survival and maintenance of cellular homeostasis by degrading and recycling intracellular components. Recent advances identify autophagy as an important regulator of myofibroblast differentiation, tissue remodeling, and fibrogenesis. In this mini-review, we provide an overview of the interactions between autophagy, ECM, and fibrosis, and emphasize the molecular mechanisms involved in myofibroblast differentiation. We also describe the emerging concept of secretory autophagy as a new avenue for intercellular communication at the site of tissue injury and repair.
Collapse
Affiliation(s)
- Francis Migneault
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM) and Université de Montréal, Montréal, QC H2X 0A9, Canada; Canadian Donation and Transplantation Research Program, Edmonton, Alberta T6G 2E1, Canada
| | - Marie-Josée Hébert
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM) and Université de Montréal, Montréal, QC H2X 0A9, Canada; Canadian Donation and Transplantation Research Program, Edmonton, Alberta T6G 2E1, Canada; Département de médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada.
| |
Collapse
|
33
|
Abstract
Human hypertrophic scars are the result of imperfect healing of skin, which is particularly evident from the scars developing after severe burns. In contrast, mouse and rat full-thickness skin wounds heal normally without forming visible scar tissue, which reduces the suitability of rodent models for the study of skin scarring. We here provide a simple procedure to splint the edges of full-thickness rodent skin with a sutured plastic frame that prevents wound closure by granulation tissue contraction. The resulting mechanical tension in the wound bed and the lack of neo-epithelium amplify myofibroblast formation and generate hypertrophic features, not unlike those of human skin. In addition to producing scar tissue, the splint provides a reservoir that can be used for the delivery of cellular and acellular wound treatment regimen. Despite being simple and almost historical, wound splinting is a robust and reliable model to study myofibroblast biology.
Collapse
Affiliation(s)
- Dong Ok Son
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
34
|
Aujla PK, Kassiri Z. Diverse origins and activation of fibroblasts in cardiac fibrosis. Cell Signal 2020; 78:109869. [PMID: 33278559 DOI: 10.1016/j.cellsig.2020.109869] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/21/2022]
Abstract
Cardiac fibroblasts (cFBs) have emerged as a heterogenous cell population. Fibroblasts are considered the main cell source for synthesis of the extracellular matrix (ECM) and as such a dysregulation in cFB function, activity, or viability can lead to disrupted ECM structure or fibrosis. Fibrosis can be initiated in response to different injuries and stimuli, and can be reparative (beneficial) or reactive (damaging). FBs need to be activated to myofibroblasts (MyoFBs) which have augmented capacity in synthesizing ECM proteins, causing fibrosis. In addition to the resident FBs in the myocardium, a number of other cells (pericytes, fibrocytes, mesenchymal, and hematopoietic cells) can transform into MyoFBs, further driving the fibrotic response. Multiple molecules including hormones, cytokines, and growth factors stimulate this process leading to generation of activated MyoFBs. Contribution of different cell types to cFBs and MyoFBs can result in an exponential increase in the number of MyoFBs and an accelerated pro-fibrotic response. Given the diversity of the cell sources, and the array of interconnected signalling pathways that lead to formation of MyoFBs and subsequently fibrosis, identifying a single target to limit the fibrotic response in the myocardium has been challenging. This review article will delineate the importance and relevance of fibroblast heterogeneity in mediating fibrosis in different models of heart failure and will highlight important signalling pathways implicated in myofibroblast activation.
Collapse
Affiliation(s)
- Preetinder K Aujla
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
35
|
Mariani P, Zhurakivska K, Santoro R, Laino G, Russo D, Laino L. Hereditary gingival fibromatosis associated with the missense mutation of the KCNK4 gene. Oral Surg Oral Med Oral Pathol Oral Radiol 2020; 131:e175-e182. [PMID: 32981868 DOI: 10.1016/j.oooo.2020.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/29/2020] [Accepted: 08/02/2020] [Indexed: 12/30/2022]
Abstract
Hereditary gingival fibromatosis (HGF) is a rare oral condition that may appear as an isolated entity or as part of a genetic disease or syndrome. Molecular and biochemical mechanisms that trigger this pathologic process are not completely understood. In this article, we present a rare case of hereditary gingival fibromatosis in conjunction with a syndromic phenotype, associated with a rare missense mutation of the KCNK4 gene. This mutation induces a change in the structure of the TRAAK channel belonging to the 2-pore potassium channels. The gain of function promoted by the mutation could represent the pathogenetic basis of gingival fibromatosis.
Collapse
Affiliation(s)
- Pierluigi Mariani
- Student in Oral Surgery Specialization, Multidisciplinary Department of Medical-Surgical and Odontostomatological Specialties, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Khrystyna Zhurakivska
- PhD student, Department of Clinical and Experimental Medicine, University of Foggia, Foggia
| | - Rossella Santoro
- Researcher in Odontostomatological Disciplines, Multidisciplinary Department of Medical-Surgical and Odontostomatological Specialties, University of Campania "Luigi Vanvitelli", Naples
| | - Gregorio Laino
- Full Professor of Oral and Maxillofacial Surgery, Dean, Division of Oral Surgery, Multidisciplinary Department of Medical-Surgical and Odontostomatological Specialties, University of Campania "Luigi Vanvitelli", Naples
| | - Diana Russo
- Student, Multidisciplinary Department of Medical-Surgical and Odontostomatological Specialties, University of Campania "Luigi Vanvitelli", Naples
| | - Luigi Laino
- Associate Professor of Oral Surgery, Multidisciplinary Department of Medical-Surgical and Odontostomatological Specialties, University of Campania "Luigi Vanvitelli", Naples
| |
Collapse
|
36
|
Nikoloudaki G, Snider P, Simmons O, Conway SJ, Hamilton DW. Periostin and matrix stiffness combine to regulate myofibroblast differentiation and fibronectin synthesis during palatal healing. Matrix Biol 2020; 94:31-56. [PMID: 32777343 DOI: 10.1016/j.matbio.2020.07.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/20/2022]
Abstract
Although the matricellular protein periostin is prominently upregulated in skin and gingival healing, it plays contrasting roles in myofibroblast differentiation and matrix synthesis respectively. Palatal healing is associated with scarring that can alter or restrict maxilla growth, but the expression pattern and contribution of periostin in palatal healing is unknown. Using periostin-knockout (Postn-/-) and wild-type (WT) mice, the contribution of periostin to palatal healing was investigated through 1.5 mm full-thickness excisional wounds in the hard palate. In WT mice, periostin was upregulated 6 days post-wounding, with mRNA levels peaking at day 12. Genetic deletion of periostin significantly reduced wound closure rates compared to WT mice. Absence of periostin reduced mRNA levels of pivotal genes in wound repair, including α-SMA/acta2, fibronectin and βigh3. Recruitment of fibroblasts and inflammatory cells, as visualized by immunofluorescent staining for fibroblast specific factor-1, vimentin, and macrophages markers Arginase-1 and iNOS was also impaired in Postn-/-, but not WT mice. Palatal fibroblasts isolated from the hard palate of mice were cultured on collagen gels and prefabricated silicon substrates with varying stiffness. Postn-/- fibroblasts showed a significantly reduced ability to contract a collagen gel, which was rescued by the exogenous addition of recombinant periostin. As the stiffness increased, Postn-/- fibroblasts increasingly differentiated into myofibroblasts, but not to the same degree as the WT. Pharmacological inhibition of Rac rescued the deficient myofibroblastic phenotype of Postn-/- cells. Low stiffness substrates (0.2 kPa) resulted in upregulation of fibronectin in WT cells, an effect which was significantly reduced in Postn-/- cells. Quantification of immunostaining for vinculin and integrinβ1 adhesions revealed that Periostin is required for the formation of focal and fibrillar adhesions in mPFBs. Our results suggest that periostin modulates myofibroblast differentiation and contraction via integrinβ1/RhoA pathway, and fibronectin synthesis in an ECM stiffness dependent manner in palatal healing.
Collapse
Affiliation(s)
- Georgia Nikoloudaki
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada.
| | - Paige Snider
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut, Indianapolis, IN, United States
| | - Olga Simmons
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut, Indianapolis, IN, United States
| | - Simon J Conway
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut, Indianapolis, IN, United States
| | - Douglas W Hamilton
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada; Division of Oral Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada.
| |
Collapse
|
37
|
Nikoloudaki G, Creber K, Hamilton DW. Wound healing and fibrosis: a contrasting role for periostin in skin and the oral mucosa. Am J Physiol Cell Physiol 2020; 318:C1065-C1077. [PMID: 32267719 PMCID: PMC7311745 DOI: 10.1152/ajpcell.00035.2020] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 01/08/2023]
Abstract
Both skin and oral mucosa are characterized by the presence of keratinized epithelium in direct apposition to an underlying collagen-dense connective tissue. Despite significant overlap in structure and physiological function, skin and the oral mucosa exhibit significantly different healing profiles in response to injury. The oral mucosa has a propensity for rapid restoration of barrier function with minimal underlying fibrosis, but in contrast, skin is associated with slower healing and scar formation. Modulators of cell function, matricellular proteins have been shown to play significant roles in cutaneous healing, but their role in restoration of the oral mucosa is poorly defined. As will be discussed in this review, over the last 12 years our research group has been actively investigating the role of the profibrotic matricellular protein periostin in tissue homeostasis and fibrosis, as well as healing, in both skin and gingiva. In the skin, periostin is highly expressed in fibrotic scars and is upregulated during cutaneous wound repair, where it facilitates myofibroblast differentiation. In contrast, in gingival healing, periostin regulates extracellular matrix synthesis but does not appear to be associated with the transition of mesenchymal cells to a contractile phenotype. The significance of these findings will be discussed, with a focus on periostin as a potential therapeutic to augment healing of soft tissues or suppress fibrosis.
Collapse
Affiliation(s)
- Georgia Nikoloudaki
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
| | - Kendal Creber
- School of Biomedical Engineering, University of Western Ontario, London, Ontario, Canada
| | - Douglas W Hamilton
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
- School of Biomedical Engineering, University of Western Ontario, London, Ontario, Canada
- Division of Oral Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
38
|
Gerarduzzi C, Hartmann U, Leask A, Drobetsky E. The Matrix Revolution: Matricellular Proteins and Restructuring of the Cancer Microenvironment. Cancer Res 2020; 80:2705-2717. [PMID: 32193287 DOI: 10.1158/0008-5472.can-18-2098] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/04/2019] [Accepted: 03/17/2020] [Indexed: 11/16/2022]
Abstract
The extracellular matrix (ECM) surrounding cells is indispensable for regulating their behavior. The dynamics of ECM signaling are tightly controlled throughout growth and development. During tissue remodeling, matricellular proteins (MCP) are secreted into the ECM. These factors do not serve classical structural roles, but rather regulate matrix proteins and cell-matrix interactions to influence normal cellular functions. In the tumor microenvironment, it is becoming increasingly clear that aberrantly expressed MCPs can support multiple hallmarks of carcinogenesis by interacting with various cellular components that are coupled to an array of downstream signals. Moreover, MCPs also reorganize the biomechanical properties of the ECM to accommodate metastasis and tumor colonization. This realization is stimulating new research on MCPs as reliable and accessible biomarkers in cancer, as well as effective and selective therapeutic targets.
Collapse
Affiliation(s)
- Casimiro Gerarduzzi
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada. .,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Ursula Hartmann
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Elliot Drobetsky
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
39
|
Abstract
Allergic conjunctival diseases (ACDs) are a group of ocular allergies that include allergic conjunctivitis, atopic keratoconjunctivitis, vernal keratoconjunctivitis, and giant papillary conjunctivitis. Although a large body of information exists on the pathophysiology of ACDs, this has not yet lead to the development of clear recommendations and guidelines for the diagnosis of ACDs or development of conclusive and objective diagnostic tools. Identification of objectively measurable biomarkers that represent the molecular and cellular mechanisms associated with ACDs will be an important step toward achieving these aims. This is a comprehensive review of biological markers that have the potential to become "biomarker(s)" for ACDs and aid in the classification, diagnosis, and development of new therapeutic strategies for these group of allergic conditions.
Collapse
Affiliation(s)
- Neeta Roy
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, 930 Madison Avenue, Memphis, TN 38163
| | - Shir Levanon
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, 930 Madison Avenue, Memphis, TN 38163
| | - Penny A. Asbell
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, 930 Madison Avenue, Memphis, TN 38163
| |
Collapse
|
40
|
Evdokiou A, Kanisicak O, Gierek S, Barry A, Ivey MJ, Zhang X, Bodnar RJ, Satish L. Characterization of Burn Eschar Pericytes. J Clin Med 2020; 9:jcm9020606. [PMID: 32102389 PMCID: PMC7074206 DOI: 10.3390/jcm9020606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 02/19/2020] [Indexed: 12/31/2022] Open
Abstract
Pericytes are cells that reside adjacent to microvasculature and regulate vascular function. Pericytes gained great interest in the field of wound healing and regenerative medicine due to their multipotential fate and ability to enhance angiogenesis. In burn wounds, scarring and scar contractures are the major pathologic feature and cause loss of mobility. The present study investigated the influence of burn wound environment on pericytes during wound healing. Pericytes isolated from normal skin and tangentially excised burn eschar tissues were analyzed for differences in gene and protein expression using RNA-seq., immunocytochemistry, and ELISA analyses. RNA-seq identified 443 differentially expressed genes between normal- and burn eschar-derived pericytes. Whereas, comparing normal skin pericytes to normal skin fibroblasts identified 1021 distinct genes and comparing burn eschar pericytes to normal skin fibroblasts identified 2449 differential genes. Altogether, forkhead box E1 (FOXE1), a transcription factor, was identified as a unique marker for skin pericytes. Interestingly, FOXE1 levels were significantly elevated in burn eschar pericytes compared to normal. Additionally, burn wound pericytes showed increased expression of profibrotic genes periostin, fibronectin, and endosialin and a gain in contractile function, suggesting a contribution to scarring and fibrosis. Our findings suggest that the burn wound environment promotes pericytes to differentiate into a myofibroblast-like phenotype promoting scar formation and fibrosis.
Collapse
Affiliation(s)
- Alexander Evdokiou
- Shriners Hospitals for Children, Research Department, Cincinnati, OH 45229, USA; (A.E.); (S.G.); (A.B.)
| | - Onur Kanisicak
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA; (O.K.); (M.J.I.)
| | - Stephanie Gierek
- Shriners Hospitals for Children, Research Department, Cincinnati, OH 45229, USA; (A.E.); (S.G.); (A.B.)
| | - Amanda Barry
- Shriners Hospitals for Children, Research Department, Cincinnati, OH 45229, USA; (A.E.); (S.G.); (A.B.)
| | - Malina J. Ivey
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA; (O.K.); (M.J.I.)
| | - Xiang Zhang
- Genomics, Epigenomics and Sequencing Core, University of Cincinnati, Cincinnati, OH 45267, USA;
| | - Richard J. Bodnar
- Veterans Affairs Medical Center, University Dr. C, Pittsburgh, PA 15240, USA;
| | - Latha Satish
- Shriners Hospitals for Children, Research Department, Cincinnati, OH 45229, USA; (A.E.); (S.G.); (A.B.)
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA; (O.K.); (M.J.I.)
- Correspondence: or ; Tel.: +1-513-872-6278
| |
Collapse
|
41
|
Nikoloudaki G, Brooks S, Peidl AP, Tinney D, Hamilton DW. JNK Signaling as a Key Modulator of Soft Connective Tissue Physiology, Pathology, and Healing. Int J Mol Sci 2020; 21:E1015. [PMID: 32033060 PMCID: PMC7037145 DOI: 10.3390/ijms21031015] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/27/2020] [Accepted: 01/31/2020] [Indexed: 12/20/2022] Open
Abstract
In healthy individuals, the healing of soft tissues such as skin after pathological insult or post injury follows a relatively predictable and defined series of cell and molecular processes to restore tissue architecture and function(s). Healing progresses through the phases of hemostasis, inflammation, proliferation, remodeling, and concomitant with re-epithelialization restores barrier function. Soft tissue healing is achieved through the spatiotemporal interplay of multiple different cell types including neutrophils, monocytes/macrophages, fibroblasts, endothelial cells/pericytes, and keratinocytes. Expressed in most cell types, c-Jun N-terminal kinases (JNK) are signaling molecules associated with the regulation of several cellular processes involved in soft tissue wound healing and in response to cellular stress. A member of the mitogen-activated protein kinase family (MAPK), JNKs have been implicated in the regulation of inflammatory cell phenotype, as well as fibroblast, stem/progenitor cell, and epithelial cell biology. In this review, we discuss our understanding of JNKs in the regulation of cell behaviors related to tissue injury, pathology, and wound healing of soft tissues. Using models as diverse as Drosophila, mice, rats, as well as human tissues, research is now defining important, but sometimes conflicting roles for JNKs in the regulation of multiple molecular processes in multiple different cell types central to wound healing processes. In this review, we focus specifically on the role of JNKs in the regulation of cell behavior in the healing of skin, cornea, tendon, gingiva, and dental pulp tissues. We conclude that while parallels can be drawn between some JNK activities and the control of cell behavior in healing, the roles of JNK can also be very specific modes of action depending on the tissue and the phase of healing.
Collapse
Affiliation(s)
- Georgia Nikoloudaki
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON N6A 5C1, Canada;
| | - Sarah Brooks
- Biomedical Engineering Graduate Program, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON N6A 5C1, Canada; (S.B.); (D.T.)
| | - Alexander P. Peidl
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON N6A 5C1, Canada;
| | - Dylan Tinney
- Biomedical Engineering Graduate Program, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON N6A 5C1, Canada; (S.B.); (D.T.)
| | - Douglas W. Hamilton
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON N6A 5C1, Canada;
- Biomedical Engineering Graduate Program, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON N6A 5C1, Canada; (S.B.); (D.T.)
- Division of Oral Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON N6A 5C1, Canada
| |
Collapse
|
42
|
Rodella A, Pozzobon M, Rigon M, Franchin C, Arrigoni G, Simonato M, Ghinelli E, Vedovelli L. Topical application of lyophilized and powdered human amniotic membrane promotes diabetic ulcer healing. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.wndm.2019.100171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
43
|
Izuhara K, Nunomura S, Nanri Y, Ono J, Takai M, Kawaguchi A. Periostin: An emerging biomarker for allergic diseases. Allergy 2019; 74:2116-2128. [PMID: 30964557 DOI: 10.1111/all.13814] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/28/2019] [Accepted: 03/19/2019] [Indexed: 12/29/2022]
Abstract
Periostin is a matricellular protein as well as an extracellular matrix (ECM) protein belonging to the fasciclin family. Periostin plays important roles as a matricellular protein in the setting of allergic diseases by binding to several integrins on various cells. Since periostin is induced mainly by IL-4 and IL-13, signature type 2 cytokines, and it is highly expressed in the subepithelial regions of many chronic allergic diseases, periostin has emerged as a novel biomarker reflecting type 2 inflammation in allergic diseases. It has, moreover, been revealed that periostin has characteristics different from other type 2 biomarkers such as eosinophil count and fractional exhaled nitric oxide (FeNO), reflecting fibrosis or tissue remodeling. From this, we may say that serum periostin is a "chronic" type 2 biomarker, whereas FeNO and possibly the eosinophil count are "acute" type 2 biomarkers. In contrast, it is still uncertain how we can apply periostin measurement to the use of biologics for allergic diseases. By examining the roles of periostin in allergy and the utility and potential of periostin in developing diagnostics against allergic diseases, it is hoped that in the near future, we can develop a new strategy to treat allergic patients.
Collapse
Affiliation(s)
- Kenji Izuhara
- Division of Medical Biochemistry, Department of Biomolecular Sciences Saga Medical School Saga Japan
| | - Satoshi Nunomura
- Division of Medical Biochemistry, Department of Biomolecular Sciences Saga Medical School Saga Japan
| | - Yasuhiro Nanri
- Division of Medical Biochemistry, Department of Biomolecular Sciences Saga Medical School Saga Japan
| | - Junya Ono
- Division of Medical Biochemistry, Department of Biomolecular Sciences Saga Medical School Saga Japan
- Shino‐Test Corporation Sagamihara Japan
| | - Masayuki Takai
- Division of Medical Biochemistry, Department of Biomolecular Sciences Saga Medical School Saga Japan
- Shino‐Test Corporation Sagamihara Japan
| | - Atsushi Kawaguchi
- Education and Research Center for Community Medicine Saga Medical School Saga Japan
| |
Collapse
|
44
|
Li JY, Ren KK, Zhang WJ, Xiao L, Wu HY, Liu QY, Ding T, Zhang XC, Nie WJ, Ke Y, Deng KY, Liu QW, Xin HB. Human amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress-induced skin cell apoptosis and enhancing their proliferation through activating PI3K/AKT signaling pathway. Stem Cell Res Ther 2019; 10:247. [PMID: 31399039 PMCID: PMC6688220 DOI: 10.1186/s13287-019-1366-y] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 12/20/2022] Open
Abstract
Background Increasing evidence has shown that mesenchymal stem cells (MSCs) yield a favorable therapeutic benefit for thermal burn skin wounds. Human amniotic MSCs (hAMSCs) derived from amniotic membrane have multilineage differentiation, immunosuppressive, and anti-inflammatory potential which makes them suitable for treating skin wounds. However, the exact effects of hAMSCs on the healing of thermal burn skin wounds and their potential mechanisms are not explored. Methods hAMSCs were isolated from amniotic membrane and characterized by RT-PCR, flow cytometry, immunofluorescence, and tumorigenicity test. We assessed the effects of hAMSCs and hAMSC conditional medium (CM) on wound healing in a deep second-degree burn injury model of mice. We then investigated the biological effects of hAMSCs and hAMSC-CM on the apoptosis and proliferation of heat stress-injured human keratinocytes HaCAT and dermal fibroblasts (DFL) both in vivo and in vitro. Next, we explored the underlying mechanisms by assessing PI3K/AKT and GSK3β/β-catenin signaling pathways in heat injured HaCAT and DFL cells after hAMSCs and hAMSC-CM treatments using PI3K inhibitor LY294002 and β-catenin inhibitor ICG001. Antibody array assay was used to identify the cytokines secreted by hAMSCs that may activate PI3K/AKT signaling pathway. Results Our results showed that hAMSCs expressed various markers of embryonic stem cells and mesenchymal stem cells and have low immunogenicity and no tumorigenicity. hAMSC and hAMSC-CM transplantation significantly promoted thermal burn wound healing by accelerating re-epithelialization with increased expression of CK19 and PCNA in vivo. hAMSCs and hAMSC-CM markedly inhibited heat stress-induced apoptosis in HaCAT and DFL cells in vitro through activation of PI3K/AKT signaling and promoted their proliferation by activating GSK3β/β-catenin signaling. Furthermore, we demonstrated that hAMSC-mediated activation of GSK3β/β-catenin signaling was dependent on PI3K/AKT signaling pathway. Antibody array assay showed that a panel of cytokines including PAI-1, C-GSF, periostin, and TIMP-1 delivered from hAMSCs may contribute to the improvement of the wound healing through activating PI3K/AKT signaling pathway. Conclusion Our results demonstrated that hAMSCs and hAMSC-CM efficiently cure heat stress-induced skin injury by inhibiting apoptosis of skin cells and promoting their proliferation through activating PI3K/AKT signaling pathway, suggesting that hAMSCs and hAMSC-CM may provide an alternative therapeutic approach for the treatment of skin injury. Electronic supplementary material The online version of this article (10.1186/s13287-019-1366-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jing-Yuan Li
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, No. 1299 Xuefu Road, Honggutan District, Nanchang, 330031, People's Republic of China.,School of Life and Science, Nanchang University, Nanchang, 330031, People's Republic of China
| | - Kang-Kang Ren
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, No. 1299 Xuefu Road, Honggutan District, Nanchang, 330031, People's Republic of China
| | - Wen-Jie Zhang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, No. 1299 Xuefu Road, Honggutan District, Nanchang, 330031, People's Republic of China
| | - Ling Xiao
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, No. 1299 Xuefu Road, Honggutan District, Nanchang, 330031, People's Republic of China
| | - Han-You Wu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, No. 1299 Xuefu Road, Honggutan District, Nanchang, 330031, People's Republic of China
| | - Qian-Yu Liu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, No. 1299 Xuefu Road, Honggutan District, Nanchang, 330031, People's Republic of China
| | - Ting Ding
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, No. 1299 Xuefu Road, Honggutan District, Nanchang, 330031, People's Republic of China
| | - Xiang-Cheng Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Wen-Jia Nie
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, No. 1299 Xuefu Road, Honggutan District, Nanchang, 330031, People's Republic of China
| | - Yu Ke
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, No. 1299 Xuefu Road, Honggutan District, Nanchang, 330031, People's Republic of China
| | - Ke-Yu Deng
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, No. 1299 Xuefu Road, Honggutan District, Nanchang, 330031, People's Republic of China
| | - Quan-Wen Liu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, No. 1299 Xuefu Road, Honggutan District, Nanchang, 330031, People's Republic of China.
| | - Hong-Bo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, No. 1299 Xuefu Road, Honggutan District, Nanchang, 330031, People's Republic of China. .,School of Life and Science, Nanchang University, Nanchang, 330031, People's Republic of China.
| |
Collapse
|
45
|
Lim PJ, Lindert U, Opitz L, Hausser I, Rohrbach M, Giunta C. Transcriptome Profiling of Primary Skin Fibroblasts Reveal Distinct Molecular Features Between PLOD1- and FKBP14-Kyphoscoliotic Ehlers-Danlos Syndrome. Genes (Basel) 2019; 10:E517. [PMID: 31288483 PMCID: PMC6678841 DOI: 10.3390/genes10070517] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/04/2019] [Accepted: 07/04/2019] [Indexed: 12/03/2022] Open
Abstract
Kyphoscoliotic Ehlers-Danlos Syndrome (kEDS) is a rare genetic heterogeneous disease clinically characterized by congenital muscle hypotonia, kyphoscoliosis, and joint hypermobility. kEDS is caused by biallelic pathogenic variants in either PLOD1 or FKBP14. PLOD1 encodes the lysyl hydroxylase 1 enzyme responsible for hydroxylating lysyl residues in the collagen helix, which undergo glycosylation and form crosslinks in the extracellular matrix thus contributing to collagen fibril strength. FKBP14 encodes a peptidyl-prolyl cis-trans isomerase that catalyzes collagen folding and acts as a chaperone for types III, VI, and X collagen. Despite genetic heterogeneity, affected patients with mutations in either PLOD1 or FKBP14 are clinically indistinguishable. We aim to better understand the pathomechanism of kEDS to characterize distinguishing and overlapping molecular features underlying PLOD1-kEDS and FKBP14-kEDS, and to identify novel molecular targets that may expand treatment strategies. Transcriptome profiling by RNA sequencing of patient-derived skin fibroblasts revealed differential expression of genes encoding extracellular matrix components that are unique between PLOD1-kEDS and FKBP14-kEDS. Furthermore, we identified genes involved in inner ear development, vascular remodeling, endoplasmic reticulum (ER) stress, and protein trafficking that were differentially expressed in patient fibroblasts compared to controls. Overall, our study presents the first transcriptomics data in kEDS revealing distinct molecular features between PLOD1-kEDS and FKBP14-kEDS, and serves as a tool to better understand the disease.
Collapse
Affiliation(s)
- Pei Jin Lim
- Connective Tissue Unit, Division of Metabolism and Children's Research Centre, University Children's Hospital, 8032 Zürich, Switzerland
| | - Uschi Lindert
- Connective Tissue Unit, Division of Metabolism and Children's Research Centre, University Children's Hospital, 8032 Zürich, Switzerland
| | - Lennart Opitz
- Functional Genomics Center Zurich, University of Zurich/ETH Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Ingrid Hausser
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Marianne Rohrbach
- Connective Tissue Unit, Division of Metabolism and Children's Research Centre, University Children's Hospital, 8032 Zürich, Switzerland.
| | - Cecilia Giunta
- Connective Tissue Unit, Division of Metabolism and Children's Research Centre, University Children's Hospital, 8032 Zürich, Switzerland.
| |
Collapse
|
46
|
Maeda D, Kubo T, Kiya K, Kawai K, Matsuzaki S, Kobayashi D, Fujiwara T, Katayama T, Hosokawa K. Periostin is induced by IL-4/IL-13 in dermal fibroblasts and promotes RhoA/ROCK pathway-mediated TGF-β1 secretion in abnormal scar formation. J Plast Surg Hand Surg 2019; 53:288-294. [DOI: 10.1080/2000656x.2019.1612752] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Daisuke Maeda
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tateki Kubo
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Koichiro Kiya
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kenichiro Kawai
- Department of Plastic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Shinsuke Matsuzaki
- Department of Pharmacology, Wakayama Medical University, Kimiidera, Wakayama, Japan
| | - Daichi Kobayashi
- Department of Pharmacology, Wakayama Medical University, Kimiidera, Wakayama, Japan
| | - Toshihiro Fujiwara
- Department of Plastic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Taiichi Katayama
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan
| | - Ko Hosokawa
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
47
|
Elliott CG, Wang J, Walker JT, Michelsons S, Dunmore-Buyze J, Drangova M, Leask A, Hamilton DW. Periostin and CCN2 Scaffolds Promote the Wound Healing Response in the Skin of Diabetic Mice. Tissue Eng Part A 2019; 25:1326-1339. [PMID: 30572781 DOI: 10.1089/ten.tea.2018.0268] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
IMPACT STATEMENT Nonhealing skin wounds remain a significant burden on health care systems, with diabetic patients 20 times as likely to undergo a lower extremity amputation due to impaired healing. Novel treatments that suppress the proinflammatory signature and induce the proliferative and remodeling phases are needed clinically. We demonstrate that the addition of periostin and CCN2 in a scaffold form increases closure rates of full-thickness skin wounds in diabetic mice, concomitant with enhanced angiogenesis. Our results demonstrate the efficacy of periostin- and CCN2-containing biomaterials to stimulate wound closure, which could represent a novel method for the treatment of diabetic skin wounds.
Collapse
Affiliation(s)
- Christopher G Elliott
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Jiarong Wang
- Division of Vascular Surgery, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada.,Department of Vascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - John T Walker
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Sarah Michelsons
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Joy Dunmore-Buyze
- Imaging Research Laboratories, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Maria Drangova
- Imaging Research Laboratories, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada.,Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Andrew Leask
- Division of Oral Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Douglas W Hamilton
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada.,Division of Oral Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| |
Collapse
|
48
|
Kim SS, Nikoloudaki GE, Michelsons S, Creber K, Hamilton DW. Fibronectin synthesis, but not α-smooth muscle expression, is regulated by periostin in gingival healing through FAK/JNK signaling. Sci Rep 2019; 9:2708. [PMID: 30804350 PMCID: PMC6389918 DOI: 10.1038/s41598-018-35805-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 11/09/2018] [Indexed: 01/09/2023] Open
Abstract
During skin healing, periostin facilitates myofibroblast differentiation through a β1 integrin/FAK dependent mechanism and continued expression is associated with scarring. In contrast to skin, gingival tissue does not typically scar upon injury, but the role of periostin in gingival healing has never been investigated. Using a rat gingivectomy model, we show that the gingival architecture is re-established within 14 days of wounding. Periostin mRNA levels peak at day 7 post-wounding, with persistence of periostin protein in the connective tissue through day 14. Collagen type I and lysyl oxidase mRNA levels peak at day 7 post wounding, which corresponded with the peak of fibroblast proliferation. Although α-smooth muscle actin mRNA levels increased 200-fold in the tissue, no myofibroblasts were detected in the regenerating tissue. In vitro, human gingival fibroblast adhesion on periostin, but not collagen, was inhibited by blocking β1 integrins. Fibroblasts cultured on periostin exhibited similar rates of proliferation and myofibroblast differentiation to cells cultured on collagen only. However, human gingival fibroblasts cultured in the presence of periostin exhibited significantly increased fibronectin and collagen mRNA levels. Increases in fibronectin production were attenuated by pharmacological inhibition of FAK and JNK signaling in human gingival fibroblasts. In vivo, mRNA levels for fibronectin peaked at day 3 and 7 post wounding, with protein immunoreactivity highest at day 7, suggesting periostin is a modulator of fibronectin production during gingival healing.
Collapse
Affiliation(s)
- Shawna S Kim
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada.,Dentistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Georgia E Nikoloudaki
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Sarah Michelsons
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Kendal Creber
- Department of Biomedical Engineering, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Douglas W Hamilton
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada. .,Department of Biomedical Engineering, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada. .,Division of Oral Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada. .,Dentistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada.
| |
Collapse
|
49
|
Zhong H, Li X, Zhang J, Wu X. Overexpression of periostin is positively associated with gastric cancer metastasis through promoting tumor metastasis and invasion. J Cell Biochem 2019; 120:9927-9935. [PMID: 30637809 DOI: 10.1002/jcb.28275] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 10/24/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Gastric tumors generally have a poor prognosis and molecular markers to improve early detection and predict outcomes are greatly needed. The present study reports that periostin (POSTN), a secretory protein that can alter the remodeling of the extracellular matrix, is highly expressed in gastric tumors. MATERIALS AND METHODS Gastric tissues were collected from patients at the Department of Thoracic Surgery/Huiqiao Medical Center, Nanfang Hospital, Southern Medical University. These patients provided an informed consent and were approved by the institute. Normal, cancer, and metastatic gastric tissues from lymph nodes and tissues adjacent to the tumor were collected from patients diagnosed with gastric cancer. RESULTS Periostin expression gradually increased as the risk grade of the NIH classification increased, and this was closely correlated with disease-free survival and overall survival. Compared with adjacent normal gastric mucosa tissues, protein expression of POSTN in gastric cancer tissues and metastases was significantly higher by immunohistochemistry and Western blot analysis. In addition, POSTN was upregulated in advanced gastric cancer tissues than in early gastric cancer tissues. Moreover, the ectopic expression of POSTN in the immortalized human gastric cell line could increase the metastasis and invasion of gastric cancer cells. CONCLUSION The present results could establish the significance of POSTN in driving oncogenesis and metastasis in gastric tumors, with implications for its potential use as a diagnostic or prognostic biomarker, and as a candidate therapeutic target.
Collapse
Affiliation(s)
- Hai Zhong
- Department of Thoracic Surgery, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China.,Department of Cardiothoracic Surgery, The second Hospital of Yinzhou District, Ningbo, People's Republic of China
| | - Xiang Li
- Department of Thoracic Surgery, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Junhua Zhang
- Departmentof Operating Room, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xu Wu
- Department of Thoracic Surgery, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
50
|
Morissette Martin P, Grant A, Hamilton DW, Flynn LE. Matrix composition in 3-D collagenous bioscaffolds modulates the survival and angiogenic phenotype of human chronic wound dermal fibroblasts. Acta Biomater 2019; 83:199-210. [PMID: 30385224 DOI: 10.1016/j.actbio.2018.10.042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 10/22/2018] [Accepted: 10/27/2018] [Indexed: 01/18/2023]
Abstract
There is a substantial need for new strategies to stimulate cutaneous tissue repair in the treatment of chronic wounds. To address this challenge, our team is developing modular biomaterials termed "bead foams", comprised of porous beads synthesized exclusively of extracellular matrix (ECM) and assembled into a cohesive three-dimensional (3-D) network. In the current study, bead foams were fabricated from human decellularized adipose tissue (DAT) or commercially-sourced bovine tendon collagen (COL) to explore the effects of ECM composition on human wound edge dermal fibroblasts (weDF) sourced from chronic wound tissues. The DAT and COL bead foams were shown to be structurally similar, but compositionally distinct, containing different levels of glycosaminoglycan content and collagen types IV, V, and VI. In vitro testing under conditions simulating stresses within the chronic wound microenvironment indicated that weDF survival and angiogenic marker expression were significantly enhanced in the DAT bead foams as compared to the COL bead foams. These findings were corroborated through in vivo assessment in a subcutaneous athymic mouse model. Taken together, the results demonstrate that weDF survival and paracrine function can be modulated by the matrix source applied in the design of ECM-derived scaffolds and that the DAT bead foams hold promise as cell-instructive biological wound dressings. STATEMENT OF SIGNIFICANCE: Biological wound dressings derived from the extracellular matrix (ECM) can be designed to promote the establishment of a more permissive microenvironment for healing in the treatment of chronic wounds. In the current work, we developed modular biomaterials comprised of fused networks of porous ECM-derived beads fabricated from human decellularized adipose tissue (DAT) or commercially-available bovine collagen. The bioscaffolds were designed to be structurally similar to provide a platform for investigating the effects of ECM composition on human dermal fibroblasts isolated from chronic wounds. Testing in in vitro and in vivo models demonstrated that cell survival and pro-angiogenic function were enhanced in the adipose-derived bioscaffolds, which contained higher levels of glycosaminoglycans and collagen types IV, V, and VI. Our findings support that the complex matrix composition within DAT can induce a more pro-regenerative cellular response for applications in wound healing.
Collapse
|