1
|
Guo S, Fu L, Yin C, Shao W, Sun Q, Chen L, Xia T, Wang M, Xia H. ROS-Induced Gingival Fibroblast Senescence: Implications in Exacerbating Inflammatory Responses in Periodontal Disease. Inflammation 2024; 47:1918-1935. [PMID: 38630168 DOI: 10.1007/s10753-024-02014-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/24/2024] [Accepted: 03/26/2024] [Indexed: 11/30/2024]
Abstract
Periodontal disease is the pathological outcome of the overwhelming inflammation in periodontal tissue. Cellular senescence has been associated with chronic inflammation in several diseases. However, the role of cellular senescence in the pathogenesis of periodontal disease remained unclear. This study aimed to investigate the role and the mechanism of cellular senescence in periodontal disease. Using single-cell RNA sequencing, we first found the upregulated level of cellular senescence in fibroblasts and endothelial cells from inflamed gingival tissue. Subsequently, human gingival fibroblasts isolated from healthy and inflamed gingival tissues were labeled as H-GFs and I-GFs, respectively. Compared to H-GFs, I-GFs exhibited a distinct cellular senescence phenotype, including an increased proportion of senescence-associated β-galactosidase (SA-β-gal) positive cells, enlarged cell morphology, and significant upregulation of p16INK4A expression. We further observed increased cellular reactive oxygen species (ROS) activity, mitochondrial ROS, and DNA damage of I-GFs. These phenotypes could be reversed by ROS scavenger NAC, which suggested the cause of cellular senescence in I-GFs. The migration and proliferation assay showed the decreased activity of I-GFs while the gene expression of senescence-associated secretory phenotype (SASP) factors such as IL-1β, IL-6, TGF-β, and IL-8 was all significantly increased. Finally, we found that supernatants of I-GF culture induced more neutrophil extracellular trap (NET) formation and drove macrophage polarization toward the CD86-positive M1 pro-inflammatory phenotype. Altogether, our findings implicate that, in the inflamed gingiva, human gingival fibroblasts acquire a senescent phenotype due to oxidative stress-induced DNA and mitochondrial damage, which in turn activate neutrophils and macrophages through the secretion of SASP factors.
Collapse
Affiliation(s)
- Shuling Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Liangliang Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Chenghu Yin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Wenjun Shao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Quan Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Liangwen Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Ting Xia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Min Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| | - Haibin Xia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
2
|
Polzin A, Benkhoff M, Thienel M, Barcik M, Mourikis P, Shchurovska K, Helten C, Ehreiser V, Zhe Z, von Wulffen F, Theiss A, Peri S, Cremer S, Ahlbrecht S, Zako S, Wildeis L, Al-Kassis G, Metzen D, Utz A, Hu H, Vornholz L, Pavic G, Lüsebrink E, Strecker J, Tiedt S, Cramer M, Gliem M, Ruck T, Meuth SG, Zeus T, Mayr C, Schiller HB, Simon L, Massberg S, Kelm M, Petzold T. Long-term FXa inhibition attenuates thromboinflammation after acute myocardial infarction and stroke by platelet proteome alteration. J Thromb Haemost 2024:S1538-7836(24)00641-X. [PMID: 39551435 DOI: 10.1016/j.jtha.2024.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Immediate activated factor (F)X (FXa) inhibition exerts direct antiplatelet effects in the context of arterial thrombosis but little is known about the impact of long-term therapy on platelet function in ischemic cardiovascular diseases. OBJECTIVES Therefore, we analyzed platelet-derived effects of long-term FXa inhibition in the setting of acute myocardial infarction (AMI) and stroke. METHODS We evaluated the effect of acute versus chronic FXa inhibition on thromboinflammation following AMI and stroke in mice in vivo. Mechanistically, we identified changes in platelet gene expression and proteome under chronic FXa nonvitamin K antagonist oral anticoagulant treatment and characterized its functional consequence on platelet physiology. In a prospectively recruited cohort of patients with AMI, we determined cardiovascular magnetic resonance based cardiac endpoints under FXa nonvitamin K antagonist oral anticoagulant effects on clinical endpoints in a cohort of patients with AMI. RESULTS Chronic but not acute FXa inhibition reduced cerebral and myocardial infarct size and improved cardiac function 24 hours after AMI in mice. Mechanistically, we identified an attenuated thromboinflammatory response with reduced neutrophil extracellular trap formation in mice and patient samples. Proteome and RNA expression analysis of FXa inhibitor treated patients revealed a reduction of key regulators within the membrane trafficking and secretion machinery hampering platelet α and dense granule release. Subsequent, thromboinflammatory neutrophil extracellular trap density in thrombi isolated from stroke and myocardial infarction patients was reduced. Patients with AMI treated with FXa inhibitors showed decreased infarct size after myocardial infarction compared to patients without anticoagulation treatment. CONCLUSION Long-term FXa inhibition induces antithromboinflammatory proteome signatures in platelets, improving infarct size after myocardial infarction and stroke.
Collapse
Affiliation(s)
- Amin Polzin
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital, Düsseldorf, Germany; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| | - Marcel Benkhoff
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Institute of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Manuela Thienel
- Department of Cardiology, Ludwig-Maximilians-University Hospital, Ludwig Maximilians University, Munich, Germany; Deutsches Herzzentrum der Charité (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Maike Barcik
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Mourikis
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Khrystyna Shchurovska
- Department of Cardiology, Ludwig-Maximilians-University Hospital, Ludwig Maximilians University, Munich, Germany; Deutsches Herzzentrum der Charité (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Carolin Helten
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Vincent Ehreiser
- Deutsches Herzzentrum der Charité University Hospital Berlin, Department of Cardiology, Angiology and Intensive Care Medicine, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany; Deutsches Herzzentrum der Charité (German Centre for Cardiovascular Research), Partner site Berlin, Berlin, Germany; Friede Springer, Centre of Cardiovascular Prevention at Charité, Charité University Medicine Berlin, Berlin, Germany
| | - Zhang Zhe
- Department of Cardiology, Ludwig-Maximilians-University Hospital, Ludwig Maximilians University, Munich, Germany; Deutsches Herzzentrum der Charité (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Franziska von Wulffen
- Department of Cardiology, Ludwig-Maximilians-University Hospital, Ludwig Maximilians University, Munich, Germany; Deutsches Herzzentrum der Charité (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Alexander Theiss
- Department of Cardiology, Ludwig-Maximilians-University Hospital, Ludwig Maximilians University, Munich, Germany; Deutsches Herzzentrum der Charité (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Sameera Peri
- Department of Cardiology, Ludwig-Maximilians-University Hospital, Ludwig Maximilians University, Munich, Germany; Deutsches Herzzentrum der Charité (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Sophie Cremer
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Samantha Ahlbrecht
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Saif Zako
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Laura Wildeis
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Gabrielle Al-Kassis
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Daniel Metzen
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Amelie Utz
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Hao Hu
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Lilian Vornholz
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Goran Pavic
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Enzo Lüsebrink
- Department of Cardiology, Ludwig-Maximilians-University Hospital, Ludwig Maximilians University, Munich, Germany; Deutsches Herzzentrum der Charité (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Jan Strecker
- Department of Cardiology, Ludwig-Maximilians-University Hospital, Ludwig Maximilians University, Munich, Germany
| | - Steffen Tiedt
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-University Hospital, Ludwig Maximilians University, Munich, Germany
| | - Mareike Cramer
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Gliem
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tobias Zeus
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christoph Mayr
- Helmholtz Munich, Research Unit for Precision Regenerative Medicine (PRM), Member of the German Center for Lung Research (DZL), Munich, Germany; Institute of Experimental Pneumology, Ludwig-Maximilians University Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Herbert B Schiller
- Helmholtz Munich, Research Unit for Precision Regenerative Medicine (PRM), Member of the German Center for Lung Research (DZL), Munich, Germany; Institute of Experimental Pneumology, Ludwig-Maximilians University Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Lukas Simon
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas, USA; Therapeutic Innovation Center, Baylor College of Medicine, Houston, Texas, USA
| | - Steffen Massberg
- Department of Cardiology, Ludwig-Maximilians-University Hospital, Ludwig Maximilians University, Munich, Germany; Deutsches Herzzentrum der Charité (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty and University Hospital, Düsseldorf, Germany
| | - Tobias Petzold
- Deutsches Herzzentrum der Charité University Hospital Berlin, Department of Cardiology, Angiology and Intensive Care Medicine, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany; Deutsches Herzzentrum der Charité (German Centre for Cardiovascular Research), Partner site Berlin, Berlin, Germany; Friede Springer, Centre of Cardiovascular Prevention at Charité, Charité University Medicine Berlin, Berlin, Germany.
| |
Collapse
|
3
|
Kohlhauser M, Mayrhofer M, Kamolz LP, Smolle C. An Update on Molecular Mechanisms of Scarring-A Narrative Review. Int J Mol Sci 2024; 25:11579. [PMID: 39519131 PMCID: PMC11546163 DOI: 10.3390/ijms252111579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Fibroblasts, the principal cellular mediators of connective tissue remodeling, play a crucial role in the formation of physiological and pathological scars. Understanding the intricate interplay between fibroblasts and other cellular and molecular components is essential for elucidating the underlying mechanisms driving scar formation. Hypertrophic scars, keloids and atrophic scars arise from dysregulated wound healing processes characterized by persistent inflammation, aberrant collagen deposition, and impaired extracellular matrix remodeling. Fibroblasts play a central role in the pathogenesis of such pathological scars, driving aberrant extracellular matrix remodeling, subsequently contributing to the formation of raised or depressed fibrotic lesions. The investigation of complex interactions between fibroblasts and the microenvironment is crucial for developing targeted therapeutic interventions aimed at modulating fibroblast activity and improving clinical outcomes in patients with pathological scars. Further research into the molecular pathways governing fibroblast behavior and their heterogeneity holds promise for advancing scar management strategies. This narrative review was performed to shed light on the mechanisms behind scar formation, with a special focus on the role of fibroblasts in the formation of different types of scars, providing insights into the pathophysiology of these conditions. Through the analysis of current knowledge, this review seeks to identify the key cellular and molecular mechanisms involved in fibroblast activation, collagen synthesis, and extracellular matrix remodeling in hypertrophic scar, keloid, or atrophic scar formation.
Collapse
Affiliation(s)
- Michael Kohlhauser
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Marcel Mayrhofer
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Lars-Peter Kamolz
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- COREMED—Centre for Regenerative Medicine and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
| | - Christian Smolle
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| |
Collapse
|
4
|
Yılmaz B, Emingil G, Öztürk VÖ, Atmaca H, Köse T, Kantarcı A. Gingival crevicular fluid levels of TLR-9, AIM-2, and ZBP-1 in periodontal diseases. Oral Dis 2024. [PMID: 39171523 DOI: 10.1111/odi.15108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024]
Abstract
OBJECTIVES Toll-like receptor (TLR)-9, may play a role in periodontal disease inflammation. This study measured TLR-9 and its related molecules, absence in melanoma-2 (AIM-2) and Z-DNA-binding protein-1 (ZBP-1), in gingival crevicular fluid (GCF) from patients with varying stages of periodontal disease to assess the role of pathogen-derived nucleic acids in inflammation. MATERIALS AND METHODS The study comprised 80 participants: 20 with Stage III Grade C periodontitis, 20 with Stage III Grade B periodontitis (P-Stage III-B), 19 with gingivitis, and 21 with periodontal health. Parameters including probing depth (PD), clinical attachment level (CAL), plaque index (PI), and bleeding on probing (BOP) were recorded. ELISA was used to analyze TLR-9, AIM-2, and ZBP-1 levels in GCF. Nonparametric tests were used for statistical comparisons. RESULTS The total amount of TLR-9 was higher in P-Stage III-B than in the healthy group (p < 0.05). Similarly, the gingivitis group exhibited elevated GCF TLR-9 levels compared to the healthy group (p < 0.05). GCF AIM-2 and ZBP-1 levels remained consistent across groups (p > 0.05). Significant correlations were found between GCF TLR-9 and CAL (p < 0.05), BOP (p < 0.05), PI (p < 0.01), and GCF volume (p < 0.001). CONCLUSION These findings suggested that the TLR-9-mediated inflammatory process plays a role in periodontal disease, as evidenced by the increased levels of TLR-9 in GCF.
Collapse
Affiliation(s)
- Büşra Yılmaz
- Department of Periodontology, School of Dentistry, Ege University, İzmir, Turkey
| | - Gülnur Emingil
- Department of Periodontology, School of Dentistry, Ege University, İzmir, Turkey
| | - Veli Özgen Öztürk
- Department of Periodontology, School of Dentistry, Aydın Adnan Menderes University, Aydın, Turkey
| | - Harika Atmaca
- Department of Biology, School of Science, Celal Bayar University, Manisa, Turkey
| | - Timur Köse
- Department of Biostatistics and Medical Informatics, Ege University School of Medicine, İzmir, Turkey
| | - Alpdoğan Kantarcı
- The Forsyth Institute, Cambridge, Massachusetts, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Ma X, Li J, Li M, Qi G, Wei L, Zhang D. Nets in fibrosis: Bridging innate immunity and tissue remodeling. Int Immunopharmacol 2024; 137:112516. [PMID: 38906006 DOI: 10.1016/j.intimp.2024.112516] [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: 04/02/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 06/23/2024]
Abstract
Fibrosis, a complex pathological process characterized by excessive deposition of extracellular matrix components, leads to tissue scarring and dysfunction. Emerging evidence suggests that neutrophil extracellular traps (NETs), composed of DNA, histones, and antimicrobial proteins, significantly contribute to fibrotic diseases pathogenesis. This review summarizes the process of NETs production, molecular mechanisms, and related diseases, and outlines the cellular and molecular mechanisms associated with fibrosis. Subsequently, this review comprehensively summarizes the current understanding of the intricate interplay between NETs and fibrosis across various organs, including the lung, liver, kidney, skin, and heart. The mechanisms by which NETs contribute to fibrogenesis, including their ability to promote inflammation, induce epithelial-mesenchymal transition (EMT), activate fibroblasts, deposit extracellular matrix (ECM) components, and trigger TLR4 signaling were explored. This review aimed to provide insights into the complex relationship between NETs and fibrosis via a comprehensive analysis of existing reports, offering novel perspectives for future research and therapeutic interventions.
Collapse
Affiliation(s)
- Xueni Ma
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jipin Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Muyang Li
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Guoqing Qi
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lina Wei
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Dekui Zhang
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
6
|
Jin H, Wen X, Sun R, Yu Y, Guo Z, Yang Y, Li L, Sun B. Engineered nanovesicles from activated neutrophils with enriched bactericidal proteins have molecular debridement ability and promote infectious wound healing. BURNS & TRAUMA 2024; 12:tkae018. [PMID: 38903935 PMCID: PMC11188537 DOI: 10.1093/burnst/tkae018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/22/2024] [Accepted: 03/27/2024] [Indexed: 06/22/2024]
Abstract
Background Bacterial infections pose a considerable threat to skin wounds, particularly in the case of challenging-to-treat diabetic wounds. Systemic antibiotics often struggle to penetrate deep wound tissues and topically applied antibiotics may lead to sensitization, necessitating the development of novel approaches for effectively treating germs in deep wound tissues. Neutrophils, the predominant immune cells in the bloodstream, rapidly release an abundance of molecules via degranulation upon activation, which possess the ability to directly eliminate pathogens. This study was designed to develop novel neutrophil cell engineered nanovesicles (NVs) with high production and explore their bactericidal properties and application in promoting infectious wound healing. Methods Neutrophils were isolated from peripheral blood and activated in vitro via phorbol myristate acetate (PMA) stimulation. Engineered NVs were prepared by sequentially extruding activated neutrophils followed by ultracentrifugation and were compared with neutrophil-derived exosomes in terms of morphology, size distribution and protein contents. The bactericidal effect of NVs in vitro was evaluated using the spread plate technique, LIVE/DEAD backlight bacteria assay and observation of bacterial morphology. The therapeutic effects of NVs in vivo were evaluated using wound contraction area measurements, histopathological examinations, assessments of inflammatory factors and immunochemical staining. Results Activated neutrophils stimulated with PMA in vitro promptly release a substantial amount of bactericidal proteins. NVs are similar to exosomes in terms of morphology and particle size, but they exhibit a significantly higher enrichment of bactericidal proteins. In vitro, NVs demonstrated a significant bactericidal effect, presumably mediated by the enrichment of bactericidal proteins such as lysozyme. These NVs significantly accelerated wound healing, leading to a marked reduction in bacterial load, downregulation of inflammatory factors and enhanced collagen deposition in a full-thickness infectious skin defect model. Conclusions We developed engineered NVs derived from activated neutrophils to serve as a novel debridement method targeting bacteria in deep tissues, ultimately promoting infectious wound healing.
Collapse
Affiliation(s)
- Hangfei Jin
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, located at No. 242, Guangji Road, Gusu District, Suzhou 215008, Jiangsu Province, China
| | - Xiao Wen
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, located at No. 242, Guangji Road, Gusu District, Suzhou 215008, Jiangsu Province, China
| | - Ran Sun
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, located at No. 242, Guangji Road, Gusu District, Suzhou 215008, Jiangsu Province, China
| | - Yanzhen Yu
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, located at No. 242, Guangji Road, Gusu District, Suzhou 215008, Jiangsu Province, China
| | - Zaiwen Guo
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, located at No. 242, Guangji Road, Gusu District, Suzhou 215008, Jiangsu Province, China
| | - Yunxi Yang
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, located at No. 242, Guangji Road, Gusu District, Suzhou 215008, Jiangsu Province, China
| | - Linbin Li
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, located at No. 242, Guangji Road, Gusu District, Suzhou 215008, Jiangsu Province, China
| | - Bingwei Sun
- Research Center for Neutrophil Engineering Technology, Affiliated Suzhou Hospital of Nanjing Medical University, located at No. 242, Guangji Road, Gusu District, Suzhou 215008, Jiangsu Province, China
| |
Collapse
|
7
|
Lin S, Lin R, Zhu P, Sun X, Qiu C, Zhang B, He Y, Xu Q, Zhang H. Neutrophil extracellular traps promoting fibroblast activation and aggravating limb ischemia through Wnt5a pathway. Am J Cancer Res 2024; 14:1866-1879. [PMID: 38726275 PMCID: PMC11076237 DOI: 10.62347/sqoc7984] [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: 03/03/2024] [Accepted: 03/31/2024] [Indexed: 05/12/2024] Open
Abstract
Although the formation of NETs contributes to cancer cell invasion and distant metastasis, its role in the pathological progression of limb ischemia remains unknown. This study investigated the functional significance of NETs in cell-cell crosstalk during limb ischemia. The changes of cell subsets in lower limb ischemia samples were detected by single-cell RNA sequencing. The expression of neutrophil extracellular traps (NETs) related markers in lower limb ischemia samples was detected by immunohistochemistry and Western blotting. The signaling pathway of NETs activation in fibroblasts was verified by immunofluorescence, PCR and Western blotting. Through single-cell RNA sequencing (scRNA-seq), we identified 9 distinct cell clusters, with significantly upregulated activation levels in fibroblasts and neutrophils and phenotypic transformation of smooth muscle cells (SMCs) into a proliferative state in ischemic tissue. At the same time, the interaction between fibroblasts and smooth muscle cells was significantly enhanced in ischemic tissue. NETs levels rise and fibroblast activation is induced in ischemic conditions. Mechanistically, activated fibroblasts promote smooth muscle cell proliferation through the Wnt5a pathway. In ischemic mice, inhibition of Wnt5a mitigated vascular remodeling and subsequent ischemia. These findings highlighting the role of cell-cell crosstalk in ischemia and vascular remodeling. We found that the NETs-initiated fibroblast-SMC interaction is a critical regulator of limb ischemia via Wnt5a pathway, a potential therapeutic target for the treatment.
Collapse
Affiliation(s)
- Shigang Lin
- Department of Vascular Surgery, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhou, Zhejiang, China
| | - Ruoran Lin
- Department of Vascular Surgery, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhou, Zhejiang, China
| | - Pengwei Zhu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhou, Zhejiang, China
| | - Xiaotong Sun
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhou, Zhejiang, China
| | - Chenyang Qiu
- Department of Vascular Surgery, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhou, Zhejiang, China
| | - Bohuan Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhou, Zhejiang, China
| | - Yangyan He
- Department of Vascular Surgery, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhou, Zhejiang, China
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhou, Zhejiang, China
| | - Hongkun Zhang
- Department of Vascular Surgery, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhou, Zhejiang, China
| |
Collapse
|
8
|
Liang Q, Pan F, Qiu H, Zhou X, Cai J, Luo R, Xiong Z, Yang H, Zhang L. CLC-3 regulates TGF-β/smad signaling pathway to inhibit the process of fibrosis in hypertrophic scar. Heliyon 2024; 10:e24984. [PMID: 38333829 PMCID: PMC10850413 DOI: 10.1016/j.heliyon.2024.e24984] [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: 08/15/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 02/10/2024] Open
Abstract
Objective To study the role and mechanism of chloride channel-3 (ClC-3) in the formation of hypertrophic scar by constructing ClC-3 interference vectors and examining their effects on human hypertrophic scar fibroblasts (HSFB). Methods Human HSFB and human normal skin fibroblasts (NSFB) were used in this study, and ClC-3 interference vectors were constructed to transfect cells. ClC-3 inhibitors NPPB and Tamoxifen were used to treat cells. Cell migration and the expression of TGF-β/Smad, CollagenⅠ,CollagenⅢ were examined to explore the role of ClC-3 in the formation of hypertrophic scar. Results Compared with the normal skin tissue, the positive expression of ClC-3 and TGF-β in the scar tissue was significantly increased. The relative expression of ClC-3 and TGF-β1 in HSFB cells was higher than that in NSFB cells. Interfering with the expression of CLC-3 can inhibit the migration of HSFB cells and the expression of TGF- β/Smad, CollagenⅠ/Ⅲ. The experiment of HSFB cells treated by CLC-3 inhibitors can also obtain similar results. Conclusion Inhibiting CLC-3 can reduce the formation of hypertrophic scars.
Collapse
Affiliation(s)
- Qian Liang
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxue East Road, Xixiangtang District, Nanning City, Guangxi Province, China
| | - Fuqiang Pan
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxue East Road, Xixiangtang District, Nanning City, Guangxi Province, China
| | - Houhuang Qiu
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxue East Road, Xixiangtang District, Nanning City, Guangxi Province, China
| | - Xiang Zhou
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxue East Road, Xixiangtang District, Nanning City, Guangxi Province, China
| | - Jieyun Cai
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxue East Road, Xixiangtang District, Nanning City, Guangxi Province, China
| | - Ruijin Luo
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxue East Road, Xixiangtang District, Nanning City, Guangxi Province, China
| | - Zenghui Xiong
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxue East Road, Xixiangtang District, Nanning City, Guangxi Province, China
| | - Huawei Yang
- Department of Breast Surgery, Affiliated Cancer Hospital of Guangxi Medical University, No. 71 Hedi Road, Nanning City, Guangxi Province, China
| | - Liming Zhang
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, No. 166 Daxue East Road, Xixiangtang District, Nanning City, Guangxi Province, China
| |
Collapse
|
9
|
Zheng Y, Huang Q, Zhang Y, Geng L, Wang W, Zhang H, He X, Li Q. Multimodal roles of transient receptor potential channel activation in inducing pathological tissue scarification. Front Immunol 2023; 14:1237992. [PMID: 37705977 PMCID: PMC10497121 DOI: 10.3389/fimmu.2023.1237992] [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: 06/10/2023] [Accepted: 08/15/2023] [Indexed: 09/15/2023] Open
Abstract
Transient receptor potential (TRP) channels are a class of transmembrane proteins that can sense a variety of physical/chemical stimuli, participate in the pathological processes of various diseases and have attracted increasing attention from researchers. Recent studies have shown that some TRP channels are involved in the development of pathological scarification (PS) and directly participate in PS fibrosis and re-epithelialization or indirectly activate immune cells to release cytokines and neuropeptides, which is subdivided into immune inflammation, fibrosis, pruritus and mechanical forces increased. This review elaborates on the characteristics of TRP channels, the mechanism of PS and how TRP channels mediate the development of PS, summarizes the important role of TRP channels in the different pathogenesis of PS and proposes that therapeutic strategies targeting TRP will be important for the prevention and treatment of PS. TRP channels are expected to become new targets for PS, which will make further breakthroughs and provide potential pharmacological targets and directions for the in-depth study of PS.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiang He
- Department of Dermatology, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiannan Li
- Department of Dermatology, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|