1
|
Liu Y, Zhang W, Lin N, Yang Z, Liu Y, Chen H. SPARC activates p38γ signaling to promote PFKFB3 protein stabilization and contributes to keloid fibroblast glycolysis. Inflamm Regen 2024; 44:44. [PMID: 39482755 PMCID: PMC11529245 DOI: 10.1186/s41232-024-00357-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Keloids are currently challenging to treat because they recur after resection which may affect patients' quality of life. At present, no universal consensus on treatment regimen has been established. Thus, finding new molecular mechanisms underlying keloid formation is imminent. This study aimed to explore the function of secreted protein acidic and cysteine rich (SPARC) on keloids and its behind exact mechanisms. METHODS The expression of SPARC, p38γ, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), α-SMA, and Ki67 in patients with keloid and bleomycin (BLM)-induced fibrosis mice was assessed utilizing western blot, qRT-PCR, and immunohistochemical staining. After transfected with pcDNA-SPARC, si-SPARC-1#, si-SPARC-2#, and si-p38γ, and treated with glycolytic inhibitor (2-DG) or p38 inhibitor (SB203580), CCK-8, EdU, transwell, and western blot were utilized for assessing the proliferation, migration, and collagen production of keloid fibroblasts (KFs). RESULTS SPARC, p38γ, and PFKFB3 were highly expressed in patients with keloid and BLM-induced fibrosis mice. SPARC promoted the proliferation, migration, and collagen production of KFs via inducing glycolysis. Moreover, SPARC could activate p38γ signaling to stabilize PFKFB3 protein expression in KFs. Next, we demonstrated that SPARC promoted the proliferation, migration, collagen production, and glycolysis of KFs via regulating p38γ signaling. In addition, in BLM-induced fibrosis mice, inhibition of p38γ and PFKFB3 relieved skin fibrosis. CONCLUSIONS Our findings indicated that SPARC could activate p38γ pathway to stabilize the expression of PFKFB3, and thus promote the glycolysis of KFs and the progression of keloid.
Collapse
Affiliation(s)
- Yining Liu
- Department of Burn and Plastic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, 266100, Shandong, People's Republic of China
| | - Wei Zhang
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 JingWu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Nan Lin
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 JingWu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Zelei Yang
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 JingWu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Yanxin Liu
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 JingWu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Huaxia Chen
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 JingWu Road, Jinan, 250021, Shandong, People's Republic of China.
| |
Collapse
|
2
|
Zhang J, Li S, Kuang C, Shen Y, Yu H, Chen F, Tang R, Mao S, Lv L, Qi M, Zhang J, Yuan K. CD74 + fibroblasts proliferate upon mechanical stretching to promote angiogenesis in keloids. FASEB J 2024; 38:e70103. [PMID: 39400419 DOI: 10.1096/fj.202401302r] [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: 06/09/2024] [Revised: 09/07/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
The healing of human skin wounds is susceptible to perturbation caused by excessive mechanical stretching, resulting in enlarged scars, hypertrophic scars, or even keloids in predisposed individuals. Keloids are fibro-proliferative scar tissues that extend beyond the initial wound boundary, consisting of the actively progressing periphery and the quiescent center. The stretch-associated outgrowth and enhanced angiogenesis are two features of the periphery of keloids. However, which cell population is responsible for transducing the mechanical stimulation to the progression of keloids remains unclear. Herein, through integrative analysis of single-cell RNA sequencing of keloids, we identified CD74+ fibroblasts, a previously unappreciated subset of fibroblasts with pro-angiogenic and stretch-induced proliferative capacities, as a key player in stretch-induced progression of keloids. Immunostaining of keloid cryosections depicted a predominant distribution of CD74+ fibroblasts in the periphery, interacting with the vasculature. In vitro tube formation assays on purified CD74+ fibroblasts ascertained their pro-angiogenic function. BrdU assays revealed that these cells proliferate upon stretching, through PIEZO1-mediated calcium influx and the downstream ERK and AKT signaling. Collectively, our findings propose a model wherein CD74+ fibroblasts serve as pivotal drivers of stretch-induced keloid progression, fueled by their proliferative and pro-angiogenic activities. Targeting the attributes of CD74+ fibroblasts holds promise as a therapeutic strategy for the management of keloids.
Collapse
Affiliation(s)
- Jingheng Zhang
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuyao Li
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chunmei Kuang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yunfan Shen
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Haibin Yu
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fang Chen
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ruijun Tang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Song Mao
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lu Lv
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Min Qi
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianglin Zhang
- Department of Dermatology, Shenzhen People's Hospital, Shenzhen, Guangdong, China
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Kai Yuan
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- The Biobank of Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Lee YI, Yang Y, Ham S, Shim JE, Lee SG, Lee SH, Kim TG, Lee WJ, Kim DY, Lee JH. Heterogeneity in Keloid Scars: Influence of Mechanical Stretching on Keloids Arising from Different Anatomical Sites. J Invest Dermatol 2024:S0022-202X(24)02085-2. [PMID: 39245138 DOI: 10.1016/j.jid.2024.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 09/10/2024]
Affiliation(s)
- Young In Lee
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Republic of Korea; Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul, Republic of Korea
| | - Yohan Yang
- Bioinformatics Collaboration Unit, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seoyoon Ham
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Republic of Korea; Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung Eun Shim
- Bioinformatics Collaboration Unit, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang Gyu Lee
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Republic of Korea; Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Si-Hyung Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Tae-Gyun Kim
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Republic of Korea; Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Won Jai Lee
- Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul, Republic of Korea; Department of Plastic Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Do-Young Kim
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Republic of Korea; Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Ju Hee Lee
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Republic of Korea; Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Zhang E, Yan Q, Sun Y, Li J, Chen L, Zou J, Zeng S, Jiang J, Li J. Integrative Analysis of Lactylome and Proteome of Hypertrophic Scar To Identify Pathways or Proteins Associated with Disease Development. J Proteome Res 2024; 23:3367-3382. [PMID: 39012622 DOI: 10.1021/acs.jproteome.3c00901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Lactylation (Kla), a recently discovered post-translational modification derived from lactate, plays crucial roles in various cellular processes. However, the specific influence of lactylation on the biological processes underlying hypertrophic scar formation remains unclear. In this study, we present a comprehensive profiling of the lactylome and proteome in both hypertrophic scars and adjacent normal skin tissues. A total of 1023 Kla sites originating from 338 nonhistone proteins were identified based on lactylome analysis. Proteome analysis in hypertrophic scar and adjacent skin samples revealed the identification of 2008 proteins. It is worth noting that Kla exhibits a preference for genes associated with ribosome function as well as glycolysis/gluconeogenesis in both normal skin and hypertrophic scar tissues. Furthermore, the functional enrichment analysis demonstrated that differentially lactyled proteins are primarily involved in proteoglycans, HIF-1, and AMPK signaling pathways. The combined analysis of the lactylome and proteome data highlighted a significant upregulation of 14 lactylation sites in hypertrophic scar tissues. Overall, our investigation unveiled the significant involvement of protein lactylation in the regulation of ribosome function as well as glycolysis/gluconeogenesis, potentially contributing to the formation of hypertrophic scars.
Collapse
Affiliation(s)
- Enyuan Zhang
- Department of Plastic and Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Qiyue Yan
- Department of Plastic and Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Yue Sun
- Department of Plastic and Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Jingyun Li
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Ling Chen
- Department of Plastic and Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Jijun Zou
- Department of Burns and Plastic Surgery, Children's Hospital of Nanjing Medical University, Nanjing 210008, Jiangsu, China
| | - Siqi Zeng
- Department of Plastic and Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Jingbin Jiang
- Department of Plastic and Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Jun Li
- Department of Plastic and Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| |
Collapse
|
5
|
Xiao K, Wang S, Chen W, Hu Y, Chen Z, Liu P, Zhang J, Chen B, Zhang Z, Li X. Identification of novel immune-related signatures for keloid diagnosis and treatment: insights from integrated bulk RNA-seq and scRNA-seq analysis. Hum Genomics 2024; 18:80. [PMID: 39014455 PMCID: PMC11251391 DOI: 10.1186/s40246-024-00647-z] [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: 06/01/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Keloid is a disease characterized by proliferation of fibrous tissue after the healing of skin tissue, which seriously affects the daily life of patients. However, the clinical treatment of keloids still has limitations, that is, it is not effective in controlling keloids, resulting in a high recurrence rate. Thus, it is urgent to identify new signatures to improve the diagnosis and treatment of keloids. METHOD Bulk RNA seq and scRNA seq data were downloaded from the GEO database. First, we used WGCNA and MEGENA to co-identify keloid/immune-related DEGs. Subsequently, we used three machine learning algorithms (Randomforest, SVM-RFE, and LASSO) to identify hub immune-related genes of keloid (KHIGs) and investigated the heterogeneous expression of KHIGs during fibroblast subpopulation differentiation using scRNA-seq. Finally, we used HE and Masson staining, quantitative reverse transcription-PCR, western blotting, immunohistochemical, and Immunofluorescent assay to investigate the dysregulated expression and the mechanism of retinoic acid in keloids. RESULTS In the present study, we identified PTGFR, RBP5, and LIF as KHIGs and validated their diagnostic performance. Subsequently, we constructed a novel artificial neural network molecular diagnostic model based on the transcriptome pattern of KHIGs, which is expected to break through the current dilemma faced by molecular diagnosis of keloids in the clinic. Meanwhile, the constructed IG score can also effectively predict keloid risk, which provides a new strategy for keloid prevention. Additionally, we observed that KHIGs were also heterogeneously expressed in the constructed differentiation trajectories of fibroblast subtypes, which may affect the differentiation of fibroblast subtypes and thus lead to dysregulation of the immune microenvironment in keloids. Finally, we found that retinoic acid may treat or alleviate keloids by inhibiting RBP5 to differentiate pro-inflammatory fibroblasts (PIF) to mesenchymal fibroblasts (MF), which further reduces collagen secretion. CONCLUSION In summary, the present study provides novel immune signatures (PTGFR, RBP5, and LIF) for keloid diagnosis and treatment, and identifies retinoic acid as potential anti-keloid drugs. More importantly, we provide a new perspective for understanding the interactions between different fibroblast subtypes in keloids and the remodeling of their immune microenvironment.
Collapse
Affiliation(s)
- Kui Xiao
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Sisi Wang
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Wenxin Chen
- Department of Gynaecology and Obstetrics, Hengyang Central Hospital, Hunan Normal University, Hengyang, China
| | - Yiping Hu
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Ziang Chen
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Peng Liu
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Jinli Zhang
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Bin Chen
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China.
| | - Zhi Zhang
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China.
| | - Xiaojian Li
- Department of Plastic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China.
| |
Collapse
|
6
|
Kuroda K, Kiya K, Matsuzaki S, Takamura H, Otani N, Tomita K, Kawai K, Fujiwara T, Nakai K, Onishi A, Katayama T, Kubo T. Altered actin dynamics is possibly implicated in the inhibition of mechanical stimulation-induced dermal fibroblast differentiation into myofibroblasts. Exp Dermatol 2023; 32:2012-2022. [PMID: 37724850 DOI: 10.1111/exd.14933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/23/2023] [Accepted: 09/04/2023] [Indexed: 09/21/2023]
Abstract
The formation of hypertrophic scars and keloids is strongly associated with mechanical stimulation, and myofibroblasts are known to play a major role in abnormal scar formation. Wounds in patients with neurofibromatosis type 1 (NF1) become inconspicuous and lack the tendency to form abnormal scars. We hypothesized that there would be a unique response to mechanical stimulation and subsequent scar formation in NF1. To test this hypothesis, we investigated the molecular mechanisms of differentiation into myofibroblasts in NF1-derived fibroblasts and neurofibromin-depleted fibroblasts and examined actin dynamics, which is involved in fibroblast differentiation, with a focus on the pathway linking LIMK2/cofilin to actin dynamics. In normal fibroblasts, expression of α-smooth muscle actin (α-SMA), a marker of myofibroblasts, significantly increased after mechanical stimulation, whereas in NF1-derived and neurofibromin-depleted fibroblasts, α-SMA expression did not change. Phosphorylation of cofilin and subsequent actin polymerization did not increase in NF1-derived and neurofibromin-depleted fibroblasts after mechanical stimulation. Finally, in normal fibroblasts treated with Jasplakinolide, an actin stabilizer, α-SMA expression did not change after mechanical stimulation. Therefore, when neurofibromin was dysfunctional or depleted, subsequent actin polymerization did not occur in response to mechanical stimulation, which may have led to the unchanged expression of α-SMA. We believe this molecular pathway can be a potential therapeutic target for the treatment of abnormal scars.
Collapse
Affiliation(s)
- Kazuya Kuroda
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koichiro Kiya
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinsuke Matsuzaki
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Osaka, Japan
- Department of Radiological Sciences, Faculty of Medical Science Technology, Morinomiya University of Medical Sciences, Osaka, Japan
| | - Hironori Takamura
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Naoya Otani
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koichi Tomita
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kenichiro Kawai
- Department of Plastic Surgery, Hyogo College of Medicine, Nishinomiya, Japan
| | - Toshihiro Fujiwara
- Department of Plastic Surgery, Hyogo College of Medicine, Nishinomiya, Japan
| | - Kunihiro Nakai
- Department of Plastic and Reconstructive Surgery, University of Fukui Hospital, Fukui, Japan
| | - Ayako Onishi
- Inclusive Medical Science Research Institute, Morinomiya University of Medical Sciences, Osaka, Japan
| | - Taiichi Katayama
- Department of Child Development and Molecular Brain Science, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Tateki Kubo
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
7
|
Yu W, Li L, Tan X, Liu X, Yin C, Cao J. Development and validation of risk prediction and neural network models for dilated cardiomyopathy based on WGCNA. Front Med (Lausanne) 2023; 10:1239056. [PMID: 37869159 PMCID: PMC10585101 DOI: 10.3389/fmed.2023.1239056] [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: 06/12/2023] [Accepted: 08/07/2023] [Indexed: 10/24/2023] Open
Abstract
Background Dilated cardiomyopathy (DCM) is a progressive heart condition characterized by ventricular dilatation and impaired myocardial contractility with a high mortality rate. The molecular characterization of DCM has not been determined yet. Therefore, it is crucial to discover potential biomarkers and therapeutic options for DCM. Methods The hub genes for the DCM were screened using Weighted Gene Co-expression Network Analysis (WGCNA) and three different algorithms in Cytoscape. These genes were then validated in a mouse model of doxorubicin (DOX)-induced DCM. Based on the validated hub genes, a prediction model and a neural network model were constructed and validated in a separate dataset. Finally, we assessed the diagnostic efficiency of hub genes and their relationship with immune cells. Results A total of eight hub genes were identified. Using RT-qPCR, we validated that the expression levels of five key genes (ASPN, MFAP4, PODN, HTRA1, and FAP) were considerably higher in DCM mice compared to normal mice, and this was consistent with the microarray results. Additionally, the risk prediction and neural network models constructed from these genes showed good accuracy and sensitivity in both the combined and validation datasets. These genes also demonstrated better diagnostic power, with AUC greater than 0.7 in both the combined and validation datasets. Immune cell infiltration analysis revealed differences in the abundance of most immune cells between DCM and normal samples. Conclusion The current findings indicate an underlying association between DCM and these key genes, which could serve as potential biomarkers for diagnosing and treating DCM.
Collapse
Affiliation(s)
- Wei Yu
- Chongqing Medical University, Chongqing, China
| | - Lingjiao Li
- Chongqing Medical University, Chongqing, China
| | | | - Xiaozhu Liu
- Chongqing Medical University, Chongqing, China
| | - Chengliang Yin
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Junyi Cao
- Department of Medical Quality Control, The First People’s Hospital of Zigong City, Zigong, China
| |
Collapse
|
8
|
Medzikovic L, Aryan L, Ruffenach G, Li M, Savalli N, Sun W, Sarji S, Hong J, Sharma S, Olcese R, Fishbein G, Eghbali M. Myocardial fibrosis and calcification are attenuated by microRNA-129-5p targeting Asporin and Sox9 in cardiac fibroblasts. JCI Insight 2023; 8:e168655. [PMID: 37154157 PMCID: PMC10243800 DOI: 10.1172/jci.insight.168655] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/15/2023] [Indexed: 05/10/2023] Open
Abstract
Myocardial fibrosis and calcification associate with adverse outcomes in nonischemic heart failure. Cardiac fibroblasts (CF) transition into myofibroblasts (MF) and osteogenic fibroblasts (OF) to promote myocardial fibrosis and calcification. However, common upstream mechanisms regulating both CF-to-MF transition and CF-to-OF transition remain unknown. microRNAs are promising targets to modulate CF plasticity. Our bioinformatics revealed downregulation of miR-129-5p and upregulation of its targets small leucine-rich proteoglycan Asporin (ASPN) and transcription factor SOX9 as common in mouse and human heart failure (HF). We experimentally confirmed decreased miR-129-5p and enhanced SOX9 and ASPN expression in CF in human hearts with myocardial fibrosis and calcification. miR-129-5p repressed both CF-to-MF and CF-to-OF transition in primary CF, as did knockdown of SOX9 and ASPN. Sox9 and Aspn are direct targets of miR-129-5p that inhibit downstream β-catenin expression. Chronic Angiotensin II infusion downregulated miR-129-5p in CF in WT and TCF21-lineage CF reporter mice, and it was restored by miR-129-5p mimic. Importantly, miR-129-5p mimic not only attenuated progression of myocardial fibrosis, calcification marker expression, and SOX9 and ASPN expression in CF but also restored diastolic and systolic function. Together, we demonstrate miR-129-5p/ASPN and miR-129-5p/SOX9 as potentially novel dysregulated axes in CF-to-MF and CF-to-OF transition in myocardial fibrosis and calcification and the therapeutic relevance of miR-129-5p.
Collapse
Affiliation(s)
| | - Laila Aryan
- Department of Anesthesiology & Perioperative Medicine
| | | | - Min Li
- Department of Anesthesiology & Perioperative Medicine
| | | | - Wasila Sun
- Department of Anesthesiology & Perioperative Medicine
| | - Shervin Sarji
- Department of Anesthesiology & Perioperative Medicine
| | - Jason Hong
- Department of Anesthesiology & Perioperative Medicine
- Division of Pulmonary & Critical Care Medicine
| | - Salil Sharma
- Department of Anesthesiology & Perioperative Medicine
| | - Riccardo Olcese
- Department of Anesthesiology & Perioperative Medicine
- Department of Physiology, and
| | - Gregory Fishbein
- Department of Physiology, and
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | | |
Collapse
|
9
|
Wu T, Jin Y, Chen F, Xuan X, Cao J, Liang Y, Wang Y, Zhan J, Zhao M, Huang C. Identification and characterization of bone/cartilage-associated signatures in common fibrotic skin diseases. Front Genet 2023; 14:1121728. [PMID: 37082197 PMCID: PMC10111020 DOI: 10.3389/fgene.2023.1121728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
Background: Fibrotic skin diseases are characterized by excessive accumulation of the extracellular matrix (ECM) and activation of fibroblasts, leading to a global healthcare burden. However, effective treatments of fibrotic skin diseases remain limited, and their pathological mechanisms require further investigation. This study aims to investigate the common biomarkers and therapeutic targets in two major fibrotic skin diseases, namely, keloid and systemic sclerosis (SSc), by bioinformatics analysis.Methods: The keloid (GSE92566) and SSc (GSE95065) datasets were downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified, followed by functional enrichment analysis using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). We then constructed a protein–protein interaction (PPI) network for the identification of hub genes. We explored the possibility of further functional enrichment analysis of hub genes on the Metascape, GeneMANIA, and TissueNexus platforms. Transcription factor (TF)–hub gene and miRNA–hub gene networks were established using NetworkAnalyst. We fixed GSE90051 and GSE76855 as the external validation datasets. Student’s t-test and receiver operating characteristic (ROC) curve were used for candidate hub gene validation. Hub gene expression was assessed in vitro by quantitative real-time PCR.Results: A total of 157 overlapping DEGs (ODEGs) were retrieved from the GSE92566 and GSE95065 datasets, and five hub genes (COL11A1, COL5A2, ASPN, COL10A1, and COMP) were identified and validated. Functional studies revealed that hub genes were predominantly enriched in bone/cartilage-related and collagen-related processes. FOXC1 and miR-335-5p were predicted to be master regulators at both transcriptional and post‐transcriptional levels.Conclusion: COL11A1, COL5A2, ASPN, COL10A1, and COMP may help understand the pathological mechanism of the major fibrotic skin diseases; moreover, FOXC1 and miR-355-5p could build a regulatory network in keloid and SSc.
Collapse
Affiliation(s)
- Ting Wu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Jin
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangqi Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiuyun Xuan
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juanmei Cao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Liang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqing Wang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinshan Zhan
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengjie Zhao
- Department of Dermatology, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Mengjie Zhao, ; Changzheng Huang,
| | - Changzheng Huang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Mengjie Zhao, ; Changzheng Huang,
| |
Collapse
|
10
|
Xu H, Wang Z, Yang H, Zhu J, Hu Z. Bioinformatics analysis and identification of dysregulated
POSTN
in the pathogenesis of keloid. Int Wound J 2022; 20:1700-1711. [PMID: 36517972 PMCID: PMC10088861 DOI: 10.1111/iwj.14031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022] Open
Abstract
Keloid is a benign fibro-proliferative dermal tumour formed by an abnormal scarring response to injury and characterised by excessive collagen accumulation and invasive growth. The pathophysiology of keloids is complex, and the treatment for keloids is still an unmet medical need. Here, we investigated the transcriptional gene that influences keloid development by comparing keloid, non-lesioned keloid skin and normal skin as well as keloid fibroblast and normal fibroblast (GSE83286, GSE92566, GSE44270). Based on the analysis, 146 up-regulated genes and 48 down-regulated genes were found in keloid tissue compared with normal skin and keloid no-lesioned skin. Eleven genes were further identified by overlapping the DEGs from keloid tissue described previously with DEGs in keloid fibroblast. The overlapped genes included PRR16, SFRP2, EDIL3, GERM1, POSTN, PDE3A, GALNT5, F2RL2, EYA4, ZFHX4, and AIM2. POSTN is the most crucial node in PPI network, which mainly correlate to collagen-related genes. Moreover, siRNA knockdown identified POSTN is a crucial regulatory gene that regulates keloid fibroblast migration and collagen I, collagen III expression level. In conclusion, our study identified 11 hub genes that play crucial role in keloid formation and provided insights for POSTN to be the therapeutic target for keloid through bioinformatic analysis of three datasets. Additionally, our results would support the development of future therapeutic strategies.
Collapse
Affiliation(s)
- Hailin Xu
- First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
| | - Zhiyong Wang
- First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
| | - Hao Yang
- First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
| | - Jiayuan Zhu
- First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
| | - Zhicheng Hu
- First Affiliated Hospital of Sun Yat‐sen University Guangzhou China
| |
Collapse
|
11
|
Dong Y, Zhang C, Zhang Q, Li Z, Wang Y, Yan J, Wu G, Qiu L, Zhu Z, Wang B, Gu H, Zhang Y. Identification of nanoparticle-mediated siRNA-ASPN as a key gene target in the treatment of keloids. Front Bioeng Biotechnol 2022; 10:1025546. [PMID: 36394011 PMCID: PMC9649824 DOI: 10.3389/fbioe.2022.1025546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/17/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Keloid, also known as connective tissue hyperplasia, is a benign proliferative disorder with a global distribution. The available therapeutic interventions are steroid injections, surgical removal of keloids, radiotherapy, compression therapy, the application of cryosurgery, and many other methods. Objectives: Existing treatments or approaches for keloids may lead to similar or even larger lesions at the site of keloid excision, leading to a high recurrence rate. Therefore, this study aims at identifying a new gene-based therapy for the treatment of keloids. Methods: An ASPN-siRNA/nanoparticle combination (si-ASPN) and a negative siRNA/nanoparticle complex (NC) was developed on the basis of bioinformatics studies and used in vitro and in vivo experiments. Results: The results showed a strong correlation between the development of keloids and high expression of ASPN protein. With the expression of ASPN protein greatly reduced in keloid fibroblasts and nude mice allografts after treatment with si-ASPN, the collagen and fibroblasts were also uniform, thinner, parallel and regular. Conclusion: All the above experimental results suggest that keloid and ASPN are closely related and both fibroblast growth and metabolism of keloid are inhibited after silencing ASPN. Therefore, ASPN-siRNA delivered via nanoparticles can serve as a novel intervention therapy for the treatment of keloids.
Collapse
Affiliation(s)
- Yipeng Dong
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Chuwei Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Qingrong Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Institute of Burn Research, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zihan Li
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Yixiao Wang
- Medical School of Nantong University, Nantong, China
| | - Jun Yan
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Gujie Wu
- Medical School of Nantong University, Nantong, China
| | - Ling Qiu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Zhihan Zhu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Bolin Wang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Haiying Gu
- Institute of Analytical Chemistry for Life Science, Nantong University, Nantong, China
- School of Public Health, Nantong University, Nantong, China
- *Correspondence: Yi Zhang, ; Haiying Gu,
| | - Yi Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Yi Zhang, ; Haiying Gu,
| |
Collapse
|
12
|
Lin P, Zhang G, Peng R, Zhao M, Li H. Increased expression of bone/cartilage-associated genes and core transcription factors in keloids by RNA sequencing. Exp Dermatol 2022; 31:1586-1596. [PMID: 35730251 DOI: 10.1111/exd.14630] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 06/01/2022] [Accepted: 06/19/2022] [Indexed: 02/05/2023]
Abstract
Fibroblasts in keloids undergo cell identity transition with altered transcriptional characteristics. However, the core transcription factors driving this cellular reprogramming remain largely unknown. Here, we report the results of transcriptional profiling from 48 keloid and 24 control dermal tissues. We identified 1187 upregulated differentially expressed genes (foldchange > 2, false discovery rate < 0.05) in keloids, which were mainly enriched in extracellular matrix organization and bone/cartilage development, with significantly increased expression of bone/cartilage-associated collagens (COL5A1, COL10A1, and COL11A1) and glycoproteins (ACAN, COMP, and SPARC). Deconvolution analysis also revealed significantly increased composition of osteoblasts in keloid dermis. A total of 92 upregulated transcription factors were screened out from differentially expressed genes and mainly enriched in transcription process and skeleton development. Additional sequencing of six keloid individuals with multiple regions and intersection further narrow the list with 10 transcription factors. Finally, AEBP1, CREB3L1, RUNX2, and ZNF469 have been identified as candidate core regulators in promoting the gaining of bone/cartilage-like characteristics in keloids. RNA-sequencing of full-skin keloids consolidated the existence of these four transcription factors. Immunohistochemistry was employed to verify the expression of AEBP1, CREB3L1, RUNX2, and ZNF469 in keloid fibroblasts. In conclusion, we bioinformatically discovered the increased expression of bone/cartilage-associated genes and candidate core transcription factors in keloids. Our findings promise to provide molecular clues to develop novel therapeutic modalities against skin fibrosis.
Collapse
Affiliation(s)
- Pingping Lin
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing, China
- NMPA Key Laboratory for Quality Control and Evaluation of Cosmetics, Beijing, China
| | - Guohong Zhang
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Rui Peng
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing, China
- NMPA Key Laboratory for Quality Control and Evaluation of Cosmetics, Beijing, China
| | - Mingming Zhao
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing, China
- NMPA Key Laboratory for Quality Control and Evaluation of Cosmetics, Beijing, China
| | - Hang Li
- Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China
- National Clinical Research Center for Skin and Immune Diseases, Beijing, China
- NMPA Key Laboratory for Quality Control and Evaluation of Cosmetics, Beijing, China
| |
Collapse
|
13
|
Imai Y, Mori N, Nihashi Y, Kumagai Y, Shibuya Y, Oshima J, Sasaki M, Sasaki K, Aihara Y, Sekido M, Kida YS. Therapeutic Potential of Adipose Stem Cell-Derived Conditioned Medium on Scar Contraction Model. Biomedicines 2022; 10:biomedicines10102388. [PMID: 36289649 PMCID: PMC9598573 DOI: 10.3390/biomedicines10102388] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Scars are composed of stiff collagen fibers, which contract strongly owing to the action of myofibroblasts. To explore the substances that modulate scar contracture, the fibroblast-populated collagen lattice (FPCL) model has been used. However, the molecular signature of the patient-derived FPCL model has not been verified. Here, we examined whether the patient-derived keloid FPCL model reflects scar contraction, analyzing detailed gene expression changes using comprehensive RNA sequencing and histological morphology, and revealed that these models are consistent with the changes during human scar contracture. Moreover, we examined whether conditioned media derived from adipose stem cells (ASC-CM) suppress the scar contracture of the collagen disc. Detailed time-series measurements of changes in disc area showed that the addition of ASC-CM significantly inhibited the shrinkage of collagen discs. In addition, a deep sequencing data analysis revealed that ASC-CM suppressed inflammation-related gene expression in the early phase of contraction; in the later phase, this suppression was gradually replaced by extracellular matrix (ECM)-related gene expression. These lines of data suggested the effectiveness of ASC-CM in suppressing scar contractures. Therefore, the molecular analysis of the ASC-CM actions found in this study will contribute to solving medical problems regarding pathological scarring in wound prognosis.
Collapse
Affiliation(s)
- Yukiko Imai
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Ibaraki, Japan
- Department of Plastic and Reconstructive surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Nobuhito Mori
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Ibaraki, Japan
| | - Yuma Nihashi
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Ibaraki, Japan
| | - Yutaro Kumagai
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Ibaraki, Japan
| | - Yoichiro Shibuya
- Department of Plastic and Reconstructive surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Junya Oshima
- Department of Plastic and Reconstructive surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Masahiro Sasaki
- Department of Plastic and Reconstructive surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Kaoru Sasaki
- Department of Plastic and Reconstructive surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Yukiko Aihara
- Department of Plastic and Reconstructive surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Mitsuru Sekido
- Department of Plastic and Reconstructive surgery, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Yasuyuki S. Kida
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Ibaraki, Japan
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
- Correspondence: ; Tel.: +81-29-861-3000
| |
Collapse
|
14
|
Nagasaki K, Chavez M, Nagasaki A, Taylor J, Tan M, Ma M, Ralston E, Thew M, Kim DG, Somerman M, Foster B. The Bone Sialoprotein RGD Domain Modulates and Maintains Periodontal Development. J Dent Res 2022; 101:1238-1247. [PMID: 35686360 PMCID: PMC9403724 DOI: 10.1177/00220345221100794] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Bone sialoprotein (gene: Ibsp; protein: BSP) is a multifunctional extracellular matrix protein present in bone, cementum, and dentin. Accumulating evidence supports BSP as a key regulator of mineralized tissue formation via evolutionarily conserved functional domains, including a C-terminal integrin-binding Arg-Gly-Asp (RGD) domain implicated in extracellular matrix-cell signaling. Ablation of Ibsp in mice (Ibsp-/-) results in impaired bone growth and mineralization and defective osteoclastogenesis, with effects in the craniofacial region including reduced acellular cementum formation, detachment of the periodontal ligament (PDL), alveolar bone hypomineralization, and severe periodontal breakdown. We hypothesized that BSP-RGD plays an important role in cementum and alveolar bone formation and mineralization, as well as periodontal function. This hypothesis was tested by replacing the RGD motif with a nonfunctional Lys-Ala-Glu (KAE) sequence in (IbspKAE/KAE) mice and OCCM.30 murine (IbspKAE) cementoblasts. The RGD domain was not critical for acellular or cellular cementum formation in IbspKAE/KAE mice. However, PDL volume and thickness were increased, and significantly more tartrate-resistant acid phosphatase-positive osteoclasts were found on alveolar bone surfaces of IbspKAE/KAE mice versus wild type mice. PDL organization was disrupted as indicated by picrosirius red stain, second harmonic generation imaging, dynamic mechanical analysis, and decreased asporin proteoglycan localization. In vitro studies implicated RGD functions in cell migration, adhesion, and mineralization, and this was confirmed by an ossicle implant model where cells lacking BSP-RGD showed substantial defects as compared with controls. In total, the BSP-RGD domain is implicated in periodontal development, though the scale and scope of changes indicated by in vitro studies indicate that other factors may partially compensate for and reduce the phenotypic severity of mice lacking BSP-RGD in vivo.
Collapse
Affiliation(s)
- K. Nagasaki
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M.B. Chavez
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - A. Nagasaki
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J.M. Taylor
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M.H. Tan
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - M. Ma
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - E. Ralston
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M.E. Thew
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - D.-G. Kim
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - M.J. Somerman
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - B.L. Foster
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
15
|
Role of Inflammasomes in Keloids and Hypertrophic Scars-Lessons Learned from Chronic Diabetic Wounds and Skin Fibrosis. Int J Mol Sci 2022; 23:ijms23126820. [PMID: 35743263 PMCID: PMC9223684 DOI: 10.3390/ijms23126820] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 02/06/2023] Open
Abstract
Keloids and hypertrophic scars are pathological cutaneous scars. They arise from excessive wound healing, which induces chronic dermal inflammation and results in overwhelming fibroblast production of extracellular matrix. Their etiology is unclear. Inflammasomes are multiprotein complexes that are important in proinflammatory innate-immune system responses. We asked whether inflammasomes participate in pathological scarring by examining the literature on scarring, diabetic wounds (also characterized by chronic inflammation), and systemic sclerosis (also marked by fibrosis). Pathological scars are predominantly populated by anti-inflammatory M2 macrophages and recent literature hints that this could be driven by non-canonical inflammasome signaling. Diabetic-wound healing associates with inflammasome activation in immune (macrophages) and non-immune (keratinocytes) cells. Fibrotic conditions associate with inflammasome activation and inflammasome-induced transition of epithelial cells/endothelial cells/macrophages into myofibroblasts that deposit excessive extracellular matrix. Studies suggest that mechanical stimuli activate inflammasomes via the cytoskeleton and that mechanotransduction-inflammasome crosstalk is involved in fibrosis. Further research should examine (i) the roles that various inflammasome types in macrophages, (myo)fibroblasts, and other cell types play in keloid development and (ii) how mechanical stimuli interact with inflammasomes and thereby drive scar growth. Such research is likely to significantly advance our understanding of pathological scarring and aid the development of new therapeutic strategies.
Collapse
|