1
|
Cuesta-Margolles G, Schlecht-Louf G, Bachelerie F. ACKR3 in Skin Homeostasis, an Overlooked Player in the CXCR4/CXCL12 Axis. J Invest Dermatol 2025; 145:1039-1049. [PMID: 39466217 DOI: 10.1016/j.jid.2024.08.022] [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: 04/26/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 10/29/2024]
Abstract
CXCL12 and its receptor CXCR4 emerge as critical regulators within the intricate network of processes ensuring skin homeostasis. In this review, we discuss their spatial distribution and function in steady-state skin; delve into their role in acute wound healing, with emphasis on fibrotic and regenerative responses; and explore their relevance in skin responses to commensals and pathogens. Given the lack of knowledge surrounding ACKR3, the atypical receptor of CXCL12, we speculate whether and how it might be involved in the processes mentioned earlier. Is ACKR3 the (a)typical friend who enjoys missing the party, or do we need to take a closer look?
Collapse
Affiliation(s)
| | - Géraldine Schlecht-Louf
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Françoise Bachelerie
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| |
Collapse
|
2
|
Rembe JD, Garabet W, Augustin M, Dissemond J, Ibing W, Schelzig H, Stuermer EK. Immunomarker profiling in human chronic wound swabs reveals IL-1 beta/IL-1RA and CXCL8/CXCL10 ratios as potential biomarkers for wound healing, infection status and regenerative stage. J Transl Med 2025; 23:407. [PMID: 40200385 PMCID: PMC11978031 DOI: 10.1186/s12967-025-06417-2] [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: 08/30/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Chronic wounds, such as diabetic foot ulcers, venous leg ulcers, and post-surgical wound healing disorders pose a significant challenge due to prolonged healing, risk of infection, and impaired quality of life. Persistent inflammation and impaired tissue remodeling are common in these wounds. Traditional diagnostic methods, including visual inspection and microbiological cultures, offer limited insight into the wound micro-environment. Immunomarker profiling could provide a deeper understanding of the molecular mechanisms underpinning wound healing, offering potential biomarkers for infection status and healing progression. METHODS This observational, multi-center cohort study, part of the 'Wound-BIOME' project, analyzed 110 swab samples from patients with acute and chronic wounds using multiplex immunoassays. Clinical parameters such as wound type, healing status, regeneration stage, and microbial burden were recorded. Total protein concentration was assessed, and 35 key immunomarkers, including cytokines (e.g. IL- 1α, IL- 1β), chemokines (CCL2, CXCL8, CXCL10), growth factors (FGF- 2, VEGF) and matrix metalloproteinases (MMP- 7, MMP- 9, MMP- 13), were quantified. Statistical analyses were performed to correlate immunomarker levels with clinical outcomes. RESULTS Pro-inflammatory markers, such as IL- 1β, IL- 18 and chemokines like CCL2 and CXCL8, were significantly elevated in non-healing and infected wounds compared to healing wounds. The study identified two new immunomarker ratios - IL- 1β/IL- 1RA and CXCL8/CXCL10 - as potential predictors of wound healing status. The IL- 1β/IL- 1RA ratio showed the highest accuracy for distinguishing healing from non-healing wounds (AUC = 0.6837), while the CXCL8/CXCL10 ratio was most effective in identifying infection (AUC = 0.7669). CONCLUSIONS Immunomarker profiling via wound swabbing offers valuable insights into the wound healing process. Elevated levels of pro-inflammatory cytokines and MMPs are associated with chronic inflammation and impaired healing. The IL- 1β/IL- 1RA and CXCL8/CXCL10 ratios emerge as promising biomarkers to distinguish between infection and inflammation, with potential in targeted wound care. Further studies are needed to validate these findings and implement them in clinical practice.
Collapse
Affiliation(s)
- Julian-Dario Rembe
- Department for Vascular and Endovascular Surgery, University Hospital Duesseldorf (UKD), Heinrich Heine University Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany.
| | - Waseem Garabet
- Department for Vascular and Endovascular Surgery, University Hospital Duesseldorf (UKD), Heinrich Heine University Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany
| | - Matthias Augustin
- Institute for Health Services Research in Dermatology and Nursing Professions (IVDP), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Joachim Dissemond
- Department of Dermatology, Venereology and Allergology, Essen University Hospital, Essen, Germany
| | - Wiebke Ibing
- Department for Vascular and Endovascular Surgery, University Hospital Duesseldorf (UKD), Heinrich Heine University Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany
| | - Hubert Schelzig
- Department for Vascular and Endovascular Surgery, University Hospital Duesseldorf (UKD), Heinrich Heine University Duesseldorf, Moorenstrasse 5, 40225, Duesseldorf, Germany
| | - Ewa K Stuermer
- Clinic and Polyclinic for Vascular Medicine, University Heart and Vascular Center, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
3
|
Lin S, Tang L, Xu N. Research progress and strategy of FGF21 for skin wound healing. Front Med (Lausanne) 2025; 12:1510691. [PMID: 40231082 PMCID: PMC11994443 DOI: 10.3389/fmed.2025.1510691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/13/2025] [Indexed: 04/16/2025] Open
Abstract
Fibroblast Growth Factor 21 (FGF21), a pivotal member of the fibroblast growth factor family, exhibits multifaceted biological functions, including the modulation of pro-inflammatory cytokines and metabolic regulation. Recent research has revealed that in impaired skin tissues, FGF21 and its receptors are upregulated and play a significant role in accelerating the wound healing process. However, the clinical application of FGF21 is severely limited by its short in vivo half-life: this factor is often degraded by enzymes before it can exert its therapeutic effects. To address this limitation, the transdermal drug delivery system (TDDS) has emerged as an innovative approach that enables sustained drug release, significantly prolonging the therapeutic duration. Leveraging genetic recombination technology, research teams have ingeniously fused FGF21 with cell-penetrating peptides (CPPs) to construct recombinant FGF21 complexes. These novel conjugates can efficiently penetrate the epidermal barrier and achieve sustained and stable pharmacological activity through TDDS. This review systematically analyzes the potential signaling pathways by which FGF21 accelerates skin wound repair, summarizes the latest advancements in TDDS technology, explores the therapeutic potential of FGF21, and evaluates the efficacy of CPP fusion tags. The manuscript not only proposes an innovative paradigm for the application of FGF21 in skin injury treatment but also provides new insights into its use in transdermal delivery, marking a significant step toward overcoming existing clinical therapeutic challenges. From a clinical medical perspective, this innovative delivery system holds promise for addressing the bioavailability issues of traditional FGF21 therapies, offering new strategies for the clinical treatment of metabolism-related diseases and wound healing. With further research, this technology holds vast potential for clinical applications in hard-to-heal wounds such as diabetic foot ulcers and burns.
Collapse
Affiliation(s)
- Shisheng Lin
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang, China
| | - Lu Tang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Nuo Xu
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang, China
| |
Collapse
|
4
|
Liu Z, Bian X, Luo L, Björklund ÅK, Li L, Zhang L, Chen Y, Guo L, Gao J, Cao C, Wang J, He W, Xiao Y, Zhu L, Annusver K, Gopee NH, Basurto-Lozada D, Horsfall D, Bennett CL, Kasper M, Haniffa M, Sommar P, Li D, Landén NX. Spatiotemporal single-cell roadmap of human skin wound healing. Cell Stem Cell 2025; 32:479-498.e8. [PMID: 39729995 DOI: 10.1016/j.stem.2024.11.013] [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: 05/25/2024] [Revised: 09/24/2024] [Accepted: 11/22/2024] [Indexed: 12/29/2024]
Abstract
Wound healing is vital for human health, yet the details of cellular dynamics and coordination in human wound repair remain largely unexplored. To address this, we conducted single-cell multi-omics analyses on human skin wound tissues through inflammation, proliferation, and remodeling phases of wound repair from the same individuals, monitoring the cellular and molecular dynamics of human skin wound healing at an unprecedented spatiotemporal resolution. This singular roadmap reveals the cellular architecture of the wound margin and identifies FOSL1 as a critical driver of re-epithelialization. It shows that pro-inflammatory macrophages and fibroblasts sequentially support keratinocyte migration like a relay race across different healing stages. Comparison with single-cell data from venous and diabetic foot ulcers uncovers a link between failed keratinocyte migration and impaired inflammatory response in chronic wounds. Additionally, comparing human and mouse acute wound transcriptomes underscores the indispensable value of this roadmap in bridging basic research with clinical innovations.
Collapse
Affiliation(s)
- Zhuang Liu
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Xiaowei Bian
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Lihua Luo
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Åsa K Björklund
- Department of Life Science, National Bioinformatics Infrastructure Sweden, Göteborg, Sweden; Science for Life Laboratory, Chalmers University of Technology, 41296 Göteborg, Sweden
| | - Li Li
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Letian Zhang
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Yongjian Chen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Lei Guo
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Juan Gao
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Chunyan Cao
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Jiating Wang
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Wenjun He
- The first affiliated hospital of Soochow University, Department of Plastic and Burn Surgery. NO.188, Shizi Street, Suzhou, Jiangsu, China
| | - Yunting Xiao
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Liping Zhu
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China
| | - Karl Annusver
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | | | - Daniela Basurto-Lozada
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - David Horsfall
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Clare L Bennett
- Department of Haematology, University College London (UCL) Cancer Institute, London WC1E 6DD, UK
| | - Maria Kasper
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK; Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Pehr Sommar
- Department of Plastic and Reconstructive Surgery, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Dongqing Li
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042 Nanjing, China.
| | - Ning Xu Landén
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden.
| |
Collapse
|
5
|
Wang Y, Zhang Y, Ma M, Zhuang X, Lu Y, Miao L, Lu X, Cui Y, Cui W. Mechanisms underlying the involvement of peritoneal macrophages in the pathogenesis and novel therapeutic strategies for dialysis-induced peritoneal fibrosis. Front Immunol 2024; 15:1507265. [PMID: 39749340 PMCID: PMC11693514 DOI: 10.3389/fimmu.2024.1507265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Long-term exposure of the peritoneum to peritoneal dialysate results in pathophysiological changes in the anatomical organization of the peritoneum and progressive development of peritoneal fibrosis. This leads to a decline in peritoneal function and ultrafiltration failure, ultimately necessitating the discontinuation of peritoneal dialysis, severely limiting the potential for long-term maintenance. Additionally, encapsulating peritoneal sclerosis, a serious consequence of peritoneal fibrosis, resulting in patients discontinuing PD and significant mortality. The causes and mechanisms underlying peritoneal fibrosis in patients undergoing peritoneal dialysis remain unknown, with no definitive treatment available. However, abnormal activation of the immune system appears to be involved in altering the structure of the peritoneum and promoting fibrotic changes. Macrophage infiltration and polarization are key contributors to pathological injury within the peritoneum, showing a strong correlation with the epithelial-to-mesenchymal transition of mesothelial cells and driving the process of fibrosis. This article discusses the role and mechanisms underlying macrophage activation-induced peritoneal fibrosis resulting from PD by analyzing relevant literature from the past decade and provides an overview of recent therapeutic approaches targeting macrophages to treat this condition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yingchun Cui
- Department of Nephrology, Second Hospital of Jilin University,
Changchun, China
| | - Wenpeng Cui
- Department of Nephrology, Second Hospital of Jilin University,
Changchun, China
| |
Collapse
|
6
|
Efraim Y, Chen FYT, Niknezhad SV, Pham D, Cheong KN, An L, Sinada H, McNamara NA, Knox SM. Rebuilding the autoimmune-damaged corneal stroma through topical lubrication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.29.626078. [PMID: 39677756 PMCID: PMC11642755 DOI: 10.1101/2024.11.29.626078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Corneal lubrication is the most common treatment for relieving the signs and symptoms of dry eye and is considered to be largely palliative with no regenerative functions. Here we challenge this notion by demonstrating that wetting the desiccated cornea of an aqueous-deficient mouse model with the simplest form of lubrication, a saline-based solution, is sufficient to rescue the severely disrupted collagen-rich architecture of the stroma, the largest corneal compartment that is essential to transparency and vision. At the single cell level we show that stromal keratocytes responsible for maintaining stromal integrity are converted from an inflammatory state into unique reparative cell states by lubrication alone, thus revealing the extensive plasticity of these cells and the regenerative function of lubricating the surface. We further show that the generation of a reparative phenotype is due, in part, to disruption of an IL1β autocrine amplification loop promoting chronic inflammation. Thus, our study uncovers the regenerative potential of topical lubrication in dry eye and represents a paradigm shift in our understanding of its therapeutic impact.
Collapse
Affiliation(s)
- Yael Efraim
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Feeling Yu Ting Chen
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Seyyed Vahid Niknezhad
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Dylan Pham
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Ka Neng Cheong
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Luye An
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Hanan Sinada
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Nancy A. McNamara
- School of Optometry and Vision Science Graduate Program, University of California, Berkeley; Oakland, CA 94720, USA
- Department of Anatomy, University of California, San Francisco; San Francisco, CA 94143, USA
| | - Sarah M. Knox
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| |
Collapse
|
7
|
Simmons J, Gallo RL. The Central Roles of Keratinocytes in Coordinating Skin Immunity. J Invest Dermatol 2024; 144:2377-2398. [PMID: 39115524 PMCID: PMC11920965 DOI: 10.1016/j.jid.2024.06.1280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 06/06/2024] [Accepted: 06/14/2024] [Indexed: 10/25/2024]
Abstract
The function of keratinocytes (KCs) to form a barrier and produce cytokines is well-known, but recent progress has revealed many different roles for KCs in regulation of skin immunity. In this review, we provide an update on the current understanding of how KCs communicate with microbes, immunocytes, neurons, and other cells to form an effective immune barrier. We catalog the large list of genes and metabolites of KCs that participate in host defense and discuss the mechanisms of immune crosstalk, addressing how KCs simultaneously form a physical barrier, communicate with fibroblasts, and control immune signals. Overall, the signals sent and received by KCs are an exciting group of therapeutic targets to explore in the treatment of dermatologic disorders.
Collapse
Affiliation(s)
- Jared Simmons
- Department of Dermatology, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Richard L Gallo
- Department of Dermatology, School of Medicine, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
8
|
Wang M, Zan T, Fan C, Li Z, Wang D, Li Q, Zhang C. Advances in GPCR-targeted drug development in dermatology. Trends Pharmacol Sci 2024; 45:678-690. [PMID: 39060127 DOI: 10.1016/j.tips.2024.06.007] [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: 05/23/2024] [Revised: 06/16/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024]
Abstract
Achieving the efficacy and specificity of G-protein-coupled receptor (GPCR) targeting-drugs in the skin remains challenging. Understanding the molecular mechanism underlying GPCR dysfunction is crucial for developing targeted therapies. Recent advances in genetic, signal transduction, and structural studies have significantly improved our understanding of cutaneous GPCR functions in both normal and pathological states. In this review, we summarize recent discoveries of pathogenic GPCRs in dermal injuries, chronic inflammatory dermatoses, cutaneous malignancies, as well as the development of potent potential drugs. We also discuss targeting of cutaneous GPCR complexes via the transient receptor potential (TRP) channel and structure elucidation, which provide new opportunities for therapeutic targeting of GPCRs involved in skin disorders. These insights are expected to lead to more effective and specific treatments for various skin conditions.
Collapse
Affiliation(s)
- Meng Wang
- Songjiang Research Institute, Songjiang Hospital, affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Tao Zan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Chengang Fan
- Department of Orthopedics and Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Zhouxiao Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Danru Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Chao Zhang
- Department of Orthopedics and Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| |
Collapse
|
9
|
Gao J, Yu H, Pan Y, Wang X, Zhang H, Xu Y, Ma W, Zhang W, Fu L, Wang Y. Porcine cis-acting lnc-CAST positively regulates CXCL8 expression through histone H3K27ac. Vet Res 2024; 55:56. [PMID: 38715098 PMCID: PMC11077775 DOI: 10.1186/s13567-024-01296-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 02/26/2024] [Indexed: 05/12/2024] Open
Abstract
The chemokine CXCL8, also known as the neutrophil chemotactic factor, plays a crucial role in mediating inflammatory responses and managing cellular immune reactions during viral infections. Porcine reproductive and respiratory syndrome virus (PRRSV) primarily infects pulmonary alveolar macrophages (PAMs), leading to acute pulmonary infections. In this study, we explored a novel long non-coding RNA (lncRNA), termed lnc-CAST, situated within the Cxcl8 gene locus. This lncRNA was found to be highly expressed in porcine macrophages. We observed that both lnc-CAST and CXCL8 were significantly upregulated in PAMs following PRRSV infection, and after treatments with lipopolysaccharide (LPS) or lipoteichoic acid (LTA). Furthermore, we noticed a concurrent upregulation of lnc-CAST and CXCL8 expression in lungs of PRRSV-infected pigs. We then determined that lnc-CAST positively influenced CXCL8 expression in PAMs. Overexpression of lnc-CAST led to an increase in CXCL8 production, which in turn enhanced the migration of epithelial cells and the recruitment of neutrophils. Conversely, inhibiting lnc-CAST expression resulted in reduced CXCL8 production in PAMs, leading to decreased migration levels of epithelial cells and neutrophils. From a mechanistic perspective, we found that lnc-CAST, localized in the nucleus, facilitated the enrichment of histone H3K27ac in CXCL8 promoter region, thereby stimulating CXCL8 transcription in a cis-regulatory manner. In conclusion, our study underscores the pivotal critical role of lnc-CAST in regulating CXCL8 production, offering valuable insights into chemokine regulation and lung damage during PRRSV infection.
Collapse
Affiliation(s)
- Junxin Gao
- College of Veterinary Medicine, Southwest University, Chongqing, 400715, China
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Haidong Yu
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Yu Pan
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Xinrong Wang
- College of Veterinary Medicine, Southwest University, Chongqing, 400715, China
| | - He Zhang
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Yunfei Xu
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
- Chongqing Academy of Animal Science, Chongqing, 408599, China
| | - Wenjie Ma
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Wenli Zhang
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
| | - Lizhi Fu
- Chongqing Academy of Animal Science, Chongqing, 408599, China.
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China.
| | - Yue Wang
- College of Veterinary Medicine, Southwest University, Chongqing, 400715, China.
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China.
| |
Collapse
|
10
|
Park SB, Yang Y, Bang SI, Kim TS, Cho D. AESIS-1, a Rheumatoid Arthritis Therapeutic Peptide, Accelerates Wound Healing by Promoting Fibroblast Migration in a CXCR2-Dependent Manner. Int J Mol Sci 2024; 25:3937. [PMID: 38612747 PMCID: PMC11012285 DOI: 10.3390/ijms25073937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
In patients with autoimmune disorders such as rheumatoid arthritis (RA), delayed wound healing is often observed. Timely and effective wound healing is a crucial determinant of a patient's quality of life, and novel materials for skin wound repair, such as bioactive peptides, are continuously being studied and developed. One such bioactive peptide, AESIS-1, has been studied for its well-established anti-rheumatoid arthritis properties. In this study, we attempted to use the anti-RA material AESIS-1 as a therapeutic wound-healing agent based on disease-modifying antirheumatic drugs (DMARDs), which can help restore prompt wound healing. The efficacy of AESIS-1 in wound healing was assessed using a full-thickness excision model in diabetic mice; this is a well-established model for studying chronic wound repair. Initial observations revealed that mice treated with AESIS-1 exhibited significantly advanced wound repair compared with the control group. In vitro studies revealed that AESIS-1 increased the migration activity of human dermal fibroblasts (HDFs) without affecting proliferative activity. Moreover, increased HDF cell migration is mediated by upregulating chemokine receptor expression, such as that of CXC chemokine receptor 2 (CXCR2). The upregulation of CXCR2 through AESIS-1 treatment enhanced the chemotactic reactivity to CXCR2 ligands, including CXC motif ligand 8 (CXCL8). AESIS-1 directly activates the ERK and p38 mitogen-activated protein kinase (MAPK) signaling cascades, which regulate the migration and expression of CXCR2 in fibroblasts. Our results suggest that the AESIS-1 peptide is a strong wound-healing substance that increases the movement of fibroblasts and the expression of CXCR2 by turning on the ERK and p38 MAPK signaling cascades.
Collapse
Affiliation(s)
- Seung Beom Park
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Anam-dong 5-ga, Seongbuk-gu, Seoul 02841, Republic of Korea;
| | - Yoolhee Yang
- Kine Sciences, 6F, 24, Eonju-ro85gil, Gangnam-gu, Seoul 06221, Republic of Korea; (Y.Y.); (D.C.)
| | - Sa Ik Bang
- Department of Plastic Surgery, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Gangnam-gu, Seoul 06351, Republic of Korea;
| | - Tae Sung Kim
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Anam-dong 5-ga, Seongbuk-gu, Seoul 02841, Republic of Korea;
| | - Daeho Cho
- Kine Sciences, 6F, 24, Eonju-ro85gil, Gangnam-gu, Seoul 06221, Republic of Korea; (Y.Y.); (D.C.)
- Institute of Convergence Science, Korea University, Anam-dong 5-ga, Seongbuk-gu, Seoul 02841, Republic of Korea
| |
Collapse
|
11
|
Liu W. The Involvement of Cysteine-X-Cysteine Motif Chemokine Receptors in Skin Homeostasis and the Pathogenesis of Allergic Contact Dermatitis and Psoriasis. Int J Mol Sci 2024; 25:1005. [PMID: 38256077 PMCID: PMC10815665 DOI: 10.3390/ijms25021005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Members of the C-X-C motif chemokine receptor (CXCR) superfamily play central roles in initiating the innate immune response in mammalian cells by orchestrating selective cell migration and immune cell activation. With its multilayered structure, the skin, which is the largest organ in the body, performs a crucial defense function, protecting the human body from harmful environmental threats and pathogens. CXCRs contribute to primary immunological defense; these receptors are differentially expressed by different types of skin cells and act as key players in initiating downstream innate immune responses. While the initiation of inflammatory responses by CXCRs is essential for pathogen elimination and tissue healing, overactivation of these receptors can enhance T-cell-mediated autoimmune responses, resulting in excessive inflammation and the development of several skin disorders, including psoriasis, atopic dermatitis, allergic contact dermatitis, vitiligo, autoimmune diseases, and skin cancers. In summary, CXCRs serve as critical links that connect innate immunity and adaptive immunity. In this article, we present the current knowledge about the functions of CXCRs in the homeostasis function of the skin and their contributions to the pathogenesis of allergic contact dermatitis and psoriasis. Furthermore, we will examine the research progress and efficacy of therapeutic approaches that target CXCRs.
Collapse
Affiliation(s)
- Wenjie Liu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| |
Collapse
|
12
|
Lauterbach AL, Slezak AJ, Wang R, Cao S, Raczy MM, Watkins EA, Jimenez CJM, Hubbell JA. Mannose-Decorated Co-Polymer Facilitates Controlled Release of Butyrate to Accelerate Chronic Wound Healing. Adv Healthc Mater 2023; 12:e2300515. [PMID: 37503634 PMCID: PMC11468131 DOI: 10.1002/adhm.202300515] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/25/2023] [Indexed: 07/29/2023]
Abstract
Butyrate is a key bacterial metabolite that plays an important and complex role in modulation of immunity and maintenance of epithelial barriers. Its translation to clinic is limited by poor bioavailability, pungent smell, and the need for high doses, and effective delivery strategies have yet to realize clinical potential. Here, a novel polymeric delivery platform for tunable and sustainable release of butyrate consisting of a methacrylamide backbone with butyryl ester or phenyl ester side chains as well as mannosyl side chains, which is also applicable to other therapeutically relevant metabolites is reported. This platform's utility in the treatment of non-healing diabetic wounds is explored. This butyrate-containing material modulated immune cell activation in vitro and induced striking changes in the milieu of soluble cytokine and chemokine signals present within the diabetic wound microenvironment in vivo. This novel therapy shows efficacy in the treatment of non-healing wounds through the modulation of the soluble signals present within the wound, and importantly accommodates the critical temporal regulation associated with the wound healing process. Currently, the few therapies to address non-healing wounds demonstrate limited efficacy. This novel platform is positioned to address this large unmet clinical need and improve the closure of otherwise non-healing wounds.
Collapse
Affiliation(s)
| | - Anna J. Slezak
- Pritzker School of Molecular EngineeringUniversity of ChicagoChicagoIL60637USA
| | - Ruyi Wang
- Pritzker School of Molecular EngineeringUniversity of ChicagoChicagoIL60637USA
| | - Shijie Cao
- Pritzker School of Molecular EngineeringUniversity of ChicagoChicagoIL60637USA
| | - Michal M. Raczy
- Pritzker School of Molecular EngineeringUniversity of ChicagoChicagoIL60637USA
| | - Elyse A. Watkins
- Pritzker School of Molecular EngineeringUniversity of ChicagoChicagoIL60637USA
| | | | - Jeffrey A. Hubbell
- Pritzker School of Molecular EngineeringUniversity of ChicagoChicagoIL60637USA
| |
Collapse
|
13
|
Lee SH, An S, Ryu YC, Seo SH, Park S, Lee MJ, Cho SW, Choi KY. Adhesive Hydrogel Patch-Mediated Combination Drug Therapy Induces Regenerative Wound Healing through Reconstruction of Regenerative Microenvironment. Adv Healthc Mater 2023; 12:e2203094. [PMID: 36854308 DOI: 10.1002/adhm.202203094] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/22/2023] [Indexed: 03/02/2023]
Abstract
Regenerative wound healing involves the scarless wound healing as observed in fetal skin. Multiple features of regenerative wound healing have been well studied; however, the practical application of pro-regenerative materials to recapitulate the regenerative wound healing in adult skins has not yet been achieved. In this study, the authors identified that their novel pro-regenerative material, pyrogallol-functionalized hyaluronic acid (HA-PG) patches in combination with protein transduction domain-fused Dishevelled (Dvl)-binding motif (PTD-DBM), a peptide inhibiting the CXXC-type zinc finger protein 5 (CXXC5)-Dvl interaction, promoted regenerative wound healing in mice. The HA-PG patches loaded with this competitor peptide and valproic acid (VPA), a glycogen synthase kinase 3β (GSK3β) inhibitor, significantly inhibited scar formation during wound healing. The HA-PG patches with PTD-DBM and/or VPA inhibit the expression of differentiated cell markers such as α-smooth muscle actin (α-SMA) while inducing the expression of stem cell markers such as CD105 and Nestin. Moreover, Collagen III, an important factor for regenerative healing, is critically induced by the HA-PG patches with PTD-DBM and/or VPA, as also seen in VPA-treated Cxxc5-/- mouse fibroblasts. Overall, these findings suggest that the novel regeneration-promoting material can be utilized as a potential therapeutic agent to promote both wound healing and scar attenuation.
Collapse
Affiliation(s)
- Soung-Hoon Lee
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- CK Regeon Inc., Seoul, 03722, Republic of Korea
| | - Soohwan An
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yeong Chan Ryu
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seol Hwa Seo
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sohyun Park
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- CK Regeon Inc., Seoul, 03722, Republic of Korea
| | - Mi Jeong Lee
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Kang-Yell Choi
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- CK Regeon Inc., Seoul, 03722, Republic of Korea
| |
Collapse
|
14
|
Nishikori S, Yasuda J, Murata K, Takegaki J, Harada Y, Shirai Y, Fujita S. Resistance training rejuvenates aging skin by reducing circulating inflammatory factors and enhancing dermal extracellular matrices. Sci Rep 2023; 13:10214. [PMID: 37353523 PMCID: PMC10290068 DOI: 10.1038/s41598-023-37207-9] [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: 05/13/2023] [Accepted: 06/18/2023] [Indexed: 06/25/2023] Open
Abstract
Aerobic training (AT) is suggested to be an effective anti-aging strategy for skin aging. However, the respective effects of resistance training (RT) have not been studied. Therefore, we compared the effects of AT and RT on skin aging in a 16-week intervention in 61 healthy sedentary middle-aged Japanese women. Data from 56 women were available for analysis. Both interventions significantly improved skin elasticity and upper dermal structure, and RT also improved dermal thickness. After the training intervention, expression of dermal extracellular matrix-related genes was increased in normal human primary dermal fibroblasts. AT and RT had different effects on circulating levels of factors, such as cytokines, hormones in serum, and metabolites, and RT increased dermal biglycan (BGN). To our knowledge, this is the first report to show different effects of AT and RT on skin aging and identify the key factors involved in RT-induced skin rejuvenation.
Collapse
Affiliation(s)
- Shu Nishikori
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Japan
- Frontier Research Center, POLA Chemical Industries, Inc., 560 Kashio-cho, Totsuka-ku, Yokohama, Japan
| | - Jun Yasuda
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Japan
| | - Kao Murata
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Japan
| | - Junya Takegaki
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Japan
| | - Yasuko Harada
- Frontier Research Center, POLA Chemical Industries, Inc., 560 Kashio-cho, Totsuka-ku, Yokohama, Japan
| | - Yuki Shirai
- Frontier Research Center, POLA Chemical Industries, Inc., 560 Kashio-cho, Totsuka-ku, Yokohama, Japan
| | - Satoshi Fujita
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Japan.
| |
Collapse
|
15
|
Pereira B, Duque K, Ramos-Gonzalez G, Díaz-Solano D, Wittig O, Zamora M, Gledhill T, Cardier JE. Wound healing by transplantation of mesenchymal stromal cells loaded on polyethylene terephthalate scaffold: Implications for skin injury treatment. Injury 2023; 54:1071-1081. [PMID: 36801131 DOI: 10.1016/j.injury.2023.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/25/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND Several clinical studies have shown that cellular therapy based on mesenchymal stromal cells (MSCs) transplantation may accelerate wound healing. One major challenge is the delivery system used for MSCs transplantation. In this work, we evaluated the capacity of a scaffold based on polyethylene terephthalate (PET) to maintain the viability and biological functions of MSCs, in vitro. We examined the capacity of MSCs loaded on PET (MSCs/PET) to induce wound healing in an experimental model of full-thickness wound. METHODS Human MSCs were seeded and cultured on PET membranes at 37 °C for 48 h. Adhesion, viability, proliferation, migration, multipotential differentiation and chemokine production were evaluated in cultures of MSCs/PET. The possible therapeutic effect of MSCs/PET on the re-epithelialization of full thickness wounds was examined at day 3 post-wounding in C57BL/6 mice. Histological and immunohistochemical (IH) studies were performed to evaluate wound re-epithelialization and the presence of epithelial progenitor cells (EPC). As controls, wounds without treatment or treated with PET were established. RESULTS We observed MSCs adhered to PET membranes and maintained their viability, proliferation and migration. They preserved their multipotential capacity of differentiation and ability of chemokine production. MSCs/PET implants promoted an accelerated wound re-epithelialization, after three days post-wounding. It was associated with the presence of EPC Lgr6+ and K6+. DISCUSSION Our results show that MSCs/PET implants induce a rapid re-epithelialization of deep- and full-thickness wounds. MSCs/PET implants constitute a potential clinical therapy for treating cutaneous wounds.
Collapse
Affiliation(s)
- Betzabeth Pereira
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela; Laboratorio de Neurofarmacología Celular, Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela
| | - Kharelys Duque
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela
| | - Giselle Ramos-Gonzalez
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela
| | - Dylana Díaz-Solano
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela
| | - Olga Wittig
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela
| | - Mariela Zamora
- Departamento de Dermatologia, Hospital Militar "Dr Carlos Arvelo, Venezuela
| | - Teresa Gledhill
- Servicio de Anatomía Patológica, Hospital Vargas, Caracas 1010-A, Venezuela
| | - José E Cardier
- Unidad de Terapia Celular - Laboratorio de Patología Celular y Molecular, Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Apartado 21827, Caracas 1020-A, Venezuela.
| |
Collapse
|
16
|
Pyclik M, Durslewicz J, Papinska JA, Deshmukh US, Bagavant H. STING Agonist-Induced Skin Inflammation Is Exacerbated with Prior Systemic Innate Immune Activation. Int J Mol Sci 2023; 24:4128. [PMID: 36835537 PMCID: PMC9960435 DOI: 10.3390/ijms24044128] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Activation of the Stimulator of Interferon Genes (STING) protein has paradoxical outcomes in skin disease. STING activation exacerbates psoriatic skin disease and delays wound healing in diabetic mice, yet it also facilitates wound healing in normal mice. To address the role of localized STING activation in the skin, mice were injected subcutaneously with a STING agonist, diamidobenzimidazole STING Agonist-1 (diAbZi). The effect of a prior inflammatory stimulus on STING activation was addressed by pre-treating mice intraperitoneally with poly (I:C). The skin at the injection site was evaluated for local inflammation, histopathology, immune cell infiltration, and gene expression. Serum cytokine levels were measured to assess systemic inflammatory responses. Localized diABZI injection induced severe skin inflammation with erythema, scaling, and induration. However, the lesions were self-limiting and resolved within 6 weeks. At the peak of inflammation, the skin showed epidermal thickening, hyperkeratosis, and dermal fibrosis. Neutrophils, CD3 T cells, and F4/80 macrophages were present in the dermis and subcutaneous layers. Gene expression was consistent with increased local interferon and cytokine signaling. Interestingly, the poly (I:C)-pre-treated mice showed higher serum cytokine responses and developed worse inflammation with delayed wound resolution. Our study demonstrates that prior systemic inflammation amplifies STING-mediated inflammatory responses and skin disease.
Collapse
Affiliation(s)
- Marcelina Pyclik
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Justyna Durslewicz
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Joanna A. Papinska
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Umesh S. Deshmukh
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Harini Bagavant
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| |
Collapse
|
17
|
Apte A, Liechty KW, Zgheib C. Immunomodulatory biomaterials on chemokine signaling in wound healing. Front Pharmacol 2023; 14:1084948. [PMID: 37153787 PMCID: PMC10160628 DOI: 10.3389/fphar.2023.1084948] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
Normal wound healing occurs through a careful orchestration of cytokine and chemokine signaling in response to injury. Chemokines are a small family of chemotactic cytokines that are secreted by immune cells in response to injury and are primarily responsible for recruiting appropriate immune cell types to injured tissue at the appropriate time. Dysregulation of chemokine signaling is suspected to contribute to delayed wound healing and chronic wounds in diseased states. Various biomaterials are being used in the development of new therapeutics for wound healing and our understanding of their effects on chemokine signaling is limited. It has been shown that modifications to the physiochemical properties of biomaterials can affect the body's immune reaction. Studying these effects on chemokine expression by various tissues and cell type can help us develop novel biomaterial therapies. In this review, we summarize the current research available on both natural and synthetic biomaterials and their effects on chemokine signaling in wound healing. In our investigation, we conclude that our knowledge of chemokines is still limited and that many in fact share both pro-inflammatory and anti-inflammatory properties. The predominance of either a pro-inflammatory or anti-inflammatory profile is mostly likely dependent on timing after injury and exposure to the biomaterial. More research is needed to better understand the interaction and contribution of biomaterials to chemokine activity in wound healing and their immunomodulatory effects.
Collapse
|
18
|
Chen Z, Haus JM, Chen L, Jiang Y, Sverdlov M, DiPietro LA, Xiong N, Wu SC, Koh TJ, Minshall RD. Inhibition of CCL28/CCR10-Mediated eNOS Downregulation Improves Skin Wound Healing in the Obesity-Induced Mouse Model of Type 2 Diabetes. Diabetes 2022; 71:2166-2180. [PMID: 35899992 PMCID: PMC9501665 DOI: 10.2337/db21-1108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 07/21/2022] [Indexed: 11/13/2022]
Abstract
Chronic, nonhealing skin wounds, such as diabetic foot ulcers (DFUs), are common in patients with type 2 diabetes. Here, we investigated the role of chemokine (C-C motif) ligand 28 (CCL28) and its receptor C-C chemokine receptor type 10 (CCR10) in downregulation of endothelial nitric (NO) oxide synthase (eNOS) in association with delayed skin wound healing in the db/db mouse model of type 2 diabetes. We observed reduced eNOS expression and elevated CCL28/CCR10 levels in dorsal skin of db/db mice and subdermal leg biopsy specimens from human subjects with type 2 diabetes. Further interrogation revealed that overexpression of CCR10 reduced eNOS expression, NO bioavailability, and tube formation of human dermal microvascular endothelial cells (HDMVECs) in vitro, which was recapitulated in mouse dorsal skin. In addition, incubation of HDMVECs with CCL28 led to internalization of the CCR10/eNOS complex and colocalization with lysosome-associated membrane protein 1. Finally, topical application of myristoylated CCR10 binding domain 7 amino acid (Myr-CBD7) peptide prevented CCR10-eNOS interaction and subsequent eNOS downregulation, enhanced eNOS/NO levels, eNOS/VEGF-R2+ microvessel density, and blood perfusion, reduced inflammatory cytokine levels, and importantly, decreased wound healing time in db/db mice. Thus, endothelial cell CCR10 activation in genetically obese mice with type 2 diabetes promotes eNOS depletion and endothelial dysfunction, and targeted disruption of CCR10/eNOS interaction improves wound healing.
Collapse
Affiliation(s)
- Zhenlong Chen
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL
| | - Jacob M. Haus
- School of Kinesiology, University of Michigan, Ann Arbor, MI
| | - Lin Chen
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL
| | - Ying Jiang
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL
| | - Maria Sverdlov
- Research Resources Center, Research Histology and Tissue Imaging Collaborative, University of Illinois at Chicago, Chicago, IL
| | - Luisa A. DiPietro
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL
| | - Na Xiong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Stephanie C. Wu
- Departments of Surgery and Stem Cell and Regenerative Medicine, Center for Lower Extremity Ambulatory Research, Dr. William M. Scholl College of Podiatric Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL
| | - Timothy J. Koh
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL
| | - Richard D. Minshall
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
19
|
SerpinB7 deficiency contributes to development of psoriasis via calcium-mediated keratinocyte differentiation dysfunction. Cell Death Dis 2022; 13:635. [PMID: 35864103 PMCID: PMC9304369 DOI: 10.1038/s41419-022-05045-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 06/17/2022] [Accepted: 06/27/2022] [Indexed: 01/21/2023]
Abstract
Defective execution of proteases and protease inhibitors that mediate abnormal signaling cascades is emerging as a key contributor to skin diseases, such as psoriasis. SerpinB7 is identified as a skin-specific endogenous protease inhibitor, but the role and underlying mechanism in psoriasis are poorly understood. Here we found that SerpinB7 is highly expressed in psoriatic keratinocytes of patients and imiquimod-induced psoriatic lesions in mice. SerpinB7-/- mice showed abnormal epidermal barrier integrity and skin architecture in homeostasis, and aggravated psoriatic lesion with inhibiting terminal differentiation and increasing inflammatory cells infiltration compared to SerpinB7+/+ mice after Imiquimod treatment. Mechanistically, SerpinB7 deficiency results in excessive proliferation and impaired differentiation, as well as increased chemokines and antimicrobial peptide expression in normal human epidermal keratinocyte and mouse primary keratinocyte. Transcriptomics and proteomics results showed that the SeprinB7 deficiency affected keratinocyte differentiation and proinflammatory cytokines, possibly by affecting the calcium ion channel-related proteins. Notably, we demonstrated that SerpinB7 deficiency prevented the increase in intracellular Ca2+ influx, which was partly eliminated by the intracellular Ca2+ chelator BAPTA-AM. Our findings first described the critical role of SerpinB7 in the regulation of keratinocyte differentiation and psoriatic microenvironment mediated via keratinocytes' intracellular calcium flux, proposing a new candidate for therapeutic targets in psoriasis.
Collapse
|
20
|
Kim H, Kim DE, Han G, Lim NR, Kim EH, Jang Y, Cho H, Jang H, Kim KH, Kim SH, Yang Y. Harnessing the Natural Healing Power of Colostrum: Bovine Milk-Derived Extracellular Vesicles from Colostrum Facilitating the Transition from Inflammation to Tissue Regeneration for Accelerating Cutaneous Wound Healing. Adv Healthc Mater 2022; 11:e2102027. [PMID: 34865307 PMCID: PMC11468066 DOI: 10.1002/adhm.202102027] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/11/2021] [Indexed: 12/17/2022]
Abstract
As wound healing is an extremely complicated process, consisting of a cascade of interlocking biological events, successful wound healing requires a multifaceted approach to support appropriate and rapid transitions from the inflammatory to proliferative and remodeling phases. In this regard, here the potential use of bovine milk extracellular vesicles (EVs) to enhance wound healing is investigated. The results show that milk EVs promote fibroblast proliferation, migration, and endothelial tube formation. In particular, milk EVs derived from colostrum (Colos EVs) contain various anti-inflammatory factors facilitating the transition from inflammation to proliferation phase, as well as factors for tissue remodeling and angiogenesis. In an excisional wound mouse model, Colos EVs promote re-epithelialization, activate angiogenesis, and enhance extracellular matrix maturation. Interestingly, Colos EVs are further found to be quite resistant to freeze-drying procedures, maintaining their original characteristics and efficacy for wound repair after lyophilization. These findings on the superior stability and excellent activity of milk Colos EVs indicate that they hold great promise to be developed as anti-inflammatory therapeutics, especially for the treatment of cutaneous wounds.
Collapse
Affiliation(s)
- Hyosuk Kim
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Da Eun Kim
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and TechnologySeoul02792Republic of Korea
| | - Geonhee Han
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
| | - Nu Ri Lim
- Doping Control CenterKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Eun Hye Kim
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Department of Life ScienceKorea UniversitySeoul02841Republic of Korea
| | - Yeongji Jang
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Department of Life ScienceKorea UniversitySeoul02841Republic of Korea
| | - Haeun Cho
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Department of BiotechnologyKorea UniversitySeoul02841Republic of Korea
| | - Hochung Jang
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and TechnologySeoul02792Republic of Korea
| | - Ki Hun Kim
- Doping Control CenterKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Sun Hwa Kim
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Yoosoo Yang
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and TechnologySeoul02792Republic of Korea
| |
Collapse
|
21
|
Pérez LA, Leyton L, Valdivia A. Thy-1 (CD90), Integrins and Syndecan 4 are Key Regulators of Skin Wound Healing. Front Cell Dev Biol 2022; 10:810474. [PMID: 35186924 PMCID: PMC8851320 DOI: 10.3389/fcell.2022.810474] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
Acute skin wound healing is a multistage process consisting of a plethora of tightly regulated signaling events in specialized cells. The Thy-1 (CD90) glycoprotein interacts with integrins and the heparan sulfate proteoglycan syndecan 4, generating a trimolecular complex that triggers bi-directional signaling to regulate diverse aspects of the wound healing process. These proteins can act either as ligands or receptors, and they are critical for the successful progression of wound healing. The expression of Thy-1, integrins, and syndecan 4 is controlled during the healing process, and the lack of expression of any of these proteins results in delayed wound healing. Here, we review and discuss the roles and regulatory events along the stages of wound healing that support the relevance of Thy-1, integrins, and syndecan 4 as crucial regulators of skin wound healing.
Collapse
Affiliation(s)
- Leonardo A. Pérez
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
- *Correspondence: Lisette Leyton, ; Alejandra Valdivia,
| | - Alejandra Valdivia
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
- *Correspondence: Lisette Leyton, ; Alejandra Valdivia,
| |
Collapse
|
22
|
Rai V, Moellmer R, Agrawal DK. The role of CXCL8 in chronic nonhealing diabetic foot ulcers and phenotypic changes in fibroblasts: a molecular perspective. Mol Biol Rep 2022; 49:1565-1572. [PMID: 35044539 DOI: 10.1007/s11033-022-07144-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/11/2022] [Indexed: 12/23/2022]
Abstract
INTRODUCTION A persistent inflammation is perpetuated by infiltrating immune cells and cytokines secreted from these immune cells. Additionally, apoptotic keratinocytes and adipocytes in diabetes causes diabetic foot ulcer (DFU) to arrest in an inflammatory phase without progressing to the resolution phase. This leads to a nonhealing DFU and, despite advanced treatments consisting of wound debridement, off-loading the ulcer of necrotic tissue, wound dressings to keep it moist and control exudate, medication, and preventing infection, DFUs remain a clinical problem. Nonhealing DFUs pose not only an economic burden but also increased morbidity and mortality in the form of psychological stress with and increased chance of amputation, and even death. Thus, investigating the complicated underlying molecular mechanism responsible for nonhealing patterns and designing better therapeutics is warranted. This review article focuses on the role of IL-8-mediated persistent inflammation and phenotypic change of fibroblasts due to this inflammatory cascade. We have discussed various sources of interleukin (IL)-8 secretion and the possible association of IL8-fibroblast plasticity as a cause of nonhealing DFUs. MATERIAL AND METHODS A literature search on PubMed, Google Scholar, and PMC was done including the terms diabetic foot ulcer, diabetes, diabetic ulcer, chronic inflammation, interleukin 8, diabetic wound, and nonhealing diabetic foot ulcers. The articles in the English language and published in last 10 years were selected. From the pool of these, the articles describing the relationship between IL-8 and nonhealing diabetic foot ulcer and diabetic ulcer were used sorted out and used for this review article following PRISMA guidelines. CONCLUSION Increased infiltration of inflammatory immune cells, secretion of pro-inflammatory cytokines, altered keratinocyte-fibroblast function, and phenotypic changes of fibroblasts in DFUs seem to be critical to the nonhealing of DFUs. Thus, inhibiting IL-8 secretion and downstream signaling seems to be a goal of potential therapeutics.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA.
| | - Rebecca Moellmer
- Western University College of Podiatric Medicine, Pomona, CA, 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| |
Collapse
|
23
|
Savitri C, Kwon JW, Drobyshava V, Ha SS, Park K. M2 Macrophage-Derived Concentrated Conditioned Media Significantly Improves Skin Wound Healing. Tissue Eng Regen Med 2021; 19:617-628. [PMID: 34962626 PMCID: PMC9130431 DOI: 10.1007/s13770-021-00414-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Macrophages, with many different phenotypes play a major role during wound healing process, secreting the cytokines crucial to angiogenesis, cell recruitment and ECM remodeling. Therefore, macrophage-derived cytokines may be attractive therapeutic resource for wound healing. METHODS To obtain a conditioned media (CM) from macrophages, human monocyte THP-1 cells were seeded on TCP or human fibroblast-derived matrix (hFDM) and they were differentiated into M1 or M2 phenotype using distinct protocols. A combination of different substrates and macrophage phenotypes produced M1- and M2-CM or M1-hFDM- and M2-hFDM-CM, respectively. Proteome microarray determines the cytokine contents in those CMs. CMs-treated human dermal fibroblast (hDFB) was analyzed using collagen synthesis and wound scratch assay. Concentrated form of the CM (CCM), obtained by high-speed centrifugation, was administered to a murine full-thickness wound model using alginate patch, where alginate patch was incubated in the M2-CCM overnight at 4 °C before transplantation. On 14 day post-treatment, examination was carried out through H&E and Herovici staining. Keratinocyte and M2 macrophages were also evaluated via immunofluorescence staining. RESULTS Cytokine analysis of CMs found CCL1, CCL5, and G-CSF, where CCL5 is more dominant. We found increased collagen synthesis and faster wound closure in hDFB treated with M2-CM. Full-thickness wounds treated by M2-hFDM-CCM containing alginate patch showed early wound closure, larger blood vessels, increased mature collagen deposition, enhanced keratinocyte maturation and more M2-macrophage population. CONCLUSION Our study demonstrated therapeutic potential of the CM derived from M2 macrophages, where the cytokines in the CM may have played an active role for enhanced wound healing.
Collapse
Affiliation(s)
- Cininta Savitri
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea ,Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792 Republic of Korea
| | - Jae Won Kwon
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea ,Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792 Republic of Korea
| | - Valeryia Drobyshava
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea ,Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792 Republic of Korea
| | - Sang Su Ha
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea ,Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792 Republic of Korea
| |
Collapse
|
24
|
Marx C, Gardner S, Harman RM, Wagner B, Van de Walle GR. Mesenchymal stromal cell-secreted CCL2 promotes antibacterial defense mechanisms through increased antimicrobial peptide expression in keratinocytes. Stem Cells Transl Med 2021; 10:1666-1679. [PMID: 34528765 PMCID: PMC8641085 DOI: 10.1002/sctm.21-0058] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/29/2021] [Accepted: 08/11/2021] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) from both humans and horses, which represent a clinically relevant translation animal model for human cutaneous wound healing, were recently found to possess antimicrobial properties against planktonic bacteria, and in the case of equine MSCs, also against biofilms. This, together with previous findings that human and equine MSCs promote angiogenesis and wound healing, makes these cells an attractive approach to treat infected cutaneous wounds in both species. The anti-biofilm activities of equine MSC, via secretion of cysteine proteases, have only been demonstrated in vitro, thus lacking information about in vivo relevance. Moreover, the effects of the equine MSC secretome on resident skin cells have not yet been explored. The goals of this study were to (a) test the efficacy of the MSC secretome in a physiologically relevant ex vivo equine skin biofilm explant model and (b) explore the impact of the MSC secretome on the antimicrobial defense mechanisms of resident skin cells. Our salient findings were that secreted factors from equine MSCs significantly decreased viability of methicillin-resistant Staphylococcus aureus bacteria in mature biofilms in this novel skin biofilm explant model. Moreover, we demonstrated that equine MSCs secrete CCL2 that increases the antimicrobial activity of equine keratinocytes by stimulating expression of antimicrobial peptides. Collectively, these data contribute to our understanding of the MSC secretome's antimicrobial properties, both directly by killing bacteria and indirectly by stimulating immune responses of surrounding resident skin cells, thus further supporting the value of MSC secretome-based treatments for infected wounds.
Collapse
Affiliation(s)
- Charlotte Marx
- Baker Institute for Animal HealthCollege of Veterinary Medicine, Cornell UniversityIthacaNew YorkUSA
| | - Sophia Gardner
- Baker Institute for Animal HealthCollege of Veterinary Medicine, Cornell UniversityIthacaNew YorkUSA
| | - Rebecca M. Harman
- Baker Institute for Animal HealthCollege of Veterinary Medicine, Cornell UniversityIthacaNew YorkUSA
| | - Bettina Wagner
- Department of Population Medicine and Diagnostic SciencesCollege of Veterinary Medicine, Cornell UniversityIthacaNew YorkUSA
| | - Gerlinde R. Van de Walle
- Baker Institute for Animal HealthCollege of Veterinary Medicine, Cornell UniversityIthacaNew YorkUSA
| |
Collapse
|
25
|
Albuquerque-Souza E, Ishikawa KH, Amado PP, Nicoli JR, Holzhausen M, Mayer MPA. Probiotics improve re-epithelialization of scratches infected by Porphyromonas gingivalis through up-regulating CXCL8-CXCR1/CXCR2 axis. Anaerobe 2021; 72:102458. [PMID: 34547426 DOI: 10.1016/j.anaerobe.2021.102458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 10/20/2022]
Abstract
Porphyromonas gingivalis inhibits the release of CXCL8 by gingival epithelial cells and reduces their proliferation. We previously reported that Bifidocaterium sp. and Lactobacillus sp. immunomodulate gingival epithelial cells response to this periodontal pathogen, but their effects on re-epithelialization properties are still unknown. Herein we explored these activities of potential probiotics on gingival epithelial cells and clarified their mechanisms. The immortalized OBA-9 lineage was used to perform in vitro scratches. Twelve clinical isolates and commercially available strains of Bifidobacterium sp. and Lactobacillus sp. were screened. L. casei 324 m and B. pseudolongum 1191A were selected to perform mechanistic assays with P. gingivalis W83 infection and the following parameters were measured: percentage of re-epithelialization by DAPI immunofluorescence area measurement; cell number by Trypan Blue exclusion assay; CXCL8 regulation by ELISA and RT-qPCR; and expression of CXCL8 cognate receptors-CXCR1 and CXCR2 by Flow Cytometry. Complementary mechanistic assays were performed with CXCL8, in the presence or absence of the CXCR1/CXCR2 inhibitor-reparixin. L. casei 324 m and B. pseudolongum 1191A enhanced re-epithelialization/cell proliferation as well as inhibited the harmful effects of P. gingivalis W83 on these activities through an increase in the expression and release of CXCL8 and in the number of cells positive for CXCR1/CXCR2. Further, we revealed that the beneficial effects of these potential probiotics were dependent on activation of the CXCL8-CXCR1/CXCR2 axis. The current findings indicate that these potential probiotics strains may improve wound healing in the context of the periodontal tissues by a CXCL8 dependent mechanism.
Collapse
Affiliation(s)
- Emmanuel Albuquerque-Souza
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, SP, Brazil; Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| | - Karin Hitomi Ishikawa
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, SP, Brazil
| | - Pâmela Penas Amado
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| | - Jacques Robert Nicoli
- Department of Microbiology, Biological Science Institute, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marinella Holzhausen
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, SP, Brazil
| | - Marcia P A Mayer
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, SP, Brazil; Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil.
| |
Collapse
|
26
|
Controlled Release of the α-Tocopherol-Derived Metabolite α-13'-Carboxychromanol from Bacterial Nanocellulose Wound Cover Improves Wound Healing. NANOMATERIALS 2021; 11:nano11081939. [PMID: 34443772 PMCID: PMC8398652 DOI: 10.3390/nano11081939] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 01/29/2023]
Abstract
Inflammation is a hallmark of tissue remodeling during wound healing. The inflammatory response to wounds is tightly controlled and well-coordinated; dysregulation compromises wound healing and causes persistent inflammation. Topical application of natural anti-inflammatory products may improve wound healing, in particular under chronic pathological conditions. The long-chain metabolites of vitamin E (LCM) are bioactive molecules that mediate cellular effects via oxidative stress signaling as well as anti-inflammatory pathways. However, the effect of LCM on wound healing has not been investigated. We administered the α-tocopherol-derived LCMs α-13'-hydroxychromanol (α-13'-OH) and α-13'-carboxychromanol (α-13'-COOH) as well as the natural product garcinoic acid, a δ-tocotrienol derivative, in different pharmaceutical formulations directly to wounds using a splinted wound mouse model to investigate their effects on the wounds' proinflammatory microenvironment and wound healing. Garcinoic acid and, in particular, α-13'-COOH accelerated wound healing and quality of the newly formed tissue. We next loaded bacterial nanocellulose (BNC), a valuable nanomaterial used as a wound dressing with high potential for drug delivery, with α-13'-COOH. The controlled release of α-13'-COOH using BNC promoted wound healing and wound closure, mainly when a diabetic condition was induced before the injury. This study highlights the potential of α-13'-COOH combined with BNC as a potential active wound dressing for the advanced therapy of skin injuries.
Collapse
|
27
|
Go YY, Lee CM, Ju WM, Chae SW, Song JJ. Extracellular Vesicles (Secretomes) from Human Trophoblasts Promote the Regeneration of Skin Fibroblasts. Int J Mol Sci 2021; 22:ijms22136959. [PMID: 34203413 PMCID: PMC8269172 DOI: 10.3390/ijms22136959] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 01/28/2023] Open
Abstract
To date, placental trophoblasts have been of interest in the fields of obstetrics and gynecology, mainly due to their involvement in the formation of a connection between the mother and fetus that aids in placental development and fetal survival. However, the regenerative capacities of trophoblasts for application in regenerative medicine and tissue engineering are poorly understood. Here, we aim to determine the skin regeneration and anti-aging capacities of trophoblast-derived conditioned medium (TB-CM) and exosomes (TB-Exos) using human normal dermal fibroblasts (HNDFs). TB-CM and TB-Exos treatments significantly elevated the migration and proliferation potencies of HNDF cells in a dose- and time-dependent manner. When RNA sequencing (RNA-seq) was used to investigate the mechanism underlying TB-CM-induced cell migration on scratch-wounded HNDFs, the increased expression of genes associated with C-X-C motif ligand (CXCL) chemokines, toll-like receptors, and nuclear factor-kappa B (NF-κB) signaling was observed. Furthermore, treatment of intrinsically/extrinsically senescent HNDFs with TB-CM resulted in an enhanced rejuvenation of HNDFs via both protection and restoration processes. Gene expression of extracellular matrix components in the skin dermis significantly increased in TB-CM- and TB-Exos-treated HNDFs. These components are involved in the TB-CM and Exo-mediated regeneration and anti-aging of HNDFs. Thus, this study demonstrated the regenerative and anti-aging efficacies of trophoblast-derived secretomes, suggesting their potential for use in interventions for skin protection and treatment.
Collapse
Affiliation(s)
- Yoon Young Go
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, Seoul 08308, Korea; (Y.Y.G.); (C.M.L.); (W.M.J.); (S.-W.C.)
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul 08308, Korea
| | - Chan Mi Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, Seoul 08308, Korea; (Y.Y.G.); (C.M.L.); (W.M.J.); (S.-W.C.)
| | - Won Min Ju
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, Seoul 08308, Korea; (Y.Y.G.); (C.M.L.); (W.M.J.); (S.-W.C.)
| | - Sung-Won Chae
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, Seoul 08308, Korea; (Y.Y.G.); (C.M.L.); (W.M.J.); (S.-W.C.)
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul 08308, Korea
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, Seoul 08308, Korea; (Y.Y.G.); (C.M.L.); (W.M.J.); (S.-W.C.)
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul 08308, Korea
- Correspondence: ; Tel.: +82-2-2626-3186; Fax: +82-2-2626-0475
| |
Collapse
|
28
|
Paracrine interleukin-8 affects mesenchymal stem cells through the Akt pathway and enhances human umbilical vein endothelial cell proliferation and migration. Biosci Rep 2021; 41:228273. [PMID: 33843989 PMCID: PMC8493446 DOI: 10.1042/bsr20210198] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 11/17/2022] Open
Abstract
Interleukin-8 (IL-8) promotes cell homing and angiogenesis, but its effects on activating human bone marrow mesenchymal stem cells (BMSCs) and promoting angiogenesis are unclear. We used bioinformatics to predict these processes. In vitro, BMSCs were stimulated in a high-glucose (HG) environment with 50 or 100 μg/ml IL-8 was used as the IL-8 group. A total of 5 μmol/l Triciribine was added to the two IL-8 groups as the Akt inhibitor group. Cultured human umbilical vein endothelial cells (HUVECs) were cultured in BMSCs conditioned medium (CM). The changes in proliferation, apoptosis, migration ability and levels of VEGF and IL-6 in HUVECs were observed in each group. Seventy processes and 26 pathways were involved in vascular development, through which IL-8 affected BMSCs. Compared with the HG control group, HUVEC proliferation absorbance value (A value), Gap closure rate, and Transwell cell migration rate in the IL-8 50 and IL-8 100 CM groups were significantly increased (P<0.01, n=30). However, HUVEC apoptosis was significantly decreased (P<0.01, n=30). Akt and phospho-Akt (P-Akt) protein contents in lysates of BMSCs treated with IL-8, as well as VEGF and IL-6 protein contents in the supernatant of BMSCs treated with IL-8, were all highly expressed (P<0.01, n=15). These analyses confirmed that IL-8 promoted the expression of 41 core proteins in BMSCs through the PI3K Akt pathway, which could promote the proliferation and migration of vascular endothelial cells. Therefore, in an HG environment, IL-8 activated the Akt signaling pathway, promoted paracrine mechanisms of BMSCs, and improved the proliferation and migration of HUVECs.
Collapse
|
29
|
Meng L, He X, Hong Q, Qiao B, Zhang X, Wu B, Zhang X, Wei Y, Li J, Ye Z, Xiao Y. CCR4, CCR8, and P2RY14 as Prognostic Factors in Head and Neck Squamous Cell Carcinoma Are Involved in the Remodeling of the Tumor Microenvironment. Front Oncol 2021; 11:618187. [PMID: 33692955 PMCID: PMC7937936 DOI: 10.3389/fonc.2021.618187] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/06/2021] [Indexed: 12/24/2022] Open
Abstract
The tumor microenvironment (TME) plays a critical role in the initiation and progression of cancer. However, the specific mechanism of its regulation in head and neck squamous cell carcinoma (HNSCC) remains unclear. In this study, we first applied the ESTIMATE method to calculate the immune and stromal scores in patients' tumor tissues from The Cancer Genome Atlas (TCGA) database. GSE41613, GSE30784, and GSE37991 data sets from the Gene Expression Omnibus (GEO) database were recruited for further validation. Differentially expressed genes (DEGs) were identified and then analyzed by Cox regression analysis and protein-protein interaction (PPI) network construction. DEGs significantly associated with prognosis and TME will be identified as hub genes. These genes were also validated at the protein level by immunohistochemical analysis of 10 pairs of primary tumor tissues and the adjacent normal tissues from our institution. The relationship between hub genes expression and immune cell fraction estimated by CIBERSORT software was also examined. 275 DEGs were significantly associated with TME. CCR4, CCR8, and P2RY14 have then identified as hub genes by intersection Cox and PPI analysis. Further investigation revealed that the expression of CCR4, CCR8, and P2RY14 was negatively correlated with clinicopathological characteristics (clinical stage, T stage) and positively associated with survival in HNSCC patients, especially in male patients. The expression of CCR8 and P2RY14 was lower in males than in females. CCR8 and P2RY14 were differentially expressed in tumor tissues than normal tissues, and the results were validated at the protein level by immunohistochemistry experiments. Gene set enrichment analysis (GSEA) showed that the high expression groups' hub genes were mainly enriched for immune-related activities. In the low-expression groups, genes were primarily enriched in metabolic pathways. CIBERSORT results showed that the expression of these genes was all negatively correlated with the fraction of memory B cells and positively correlated with the fraction of the other four cells, including naive B cells, resting T cells CD4 memory, T cells follicular helper, and T cells regulatory (Tregs). The results suggest that CCR4, CCR8, and P2RY14 may be responsible for maintaining the immune dominance of TME, thus leading to a better prognosis.
Collapse
Affiliation(s)
- Liangliang Meng
- Medical School of Chinese PLA, Beijing, China
- Department of Radiology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
- Department of Radiology, Chinese PAP Beijing Corps Hospital, Beijing, China
| | - Xiaoxi He
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Quan Hong
- Department of Nephrology, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, China
| | - Bo Qiao
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiao Zhang
- Department of Radiology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Bin Wu
- Department of Radiology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
- Department of Radiology, Chinese PAP Beijing Corps Hospital, Beijing, China
| | - Xiaobo Zhang
- Department of Radiology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yingtian Wei
- Department of Radiology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jing Li
- Department of Radiology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yueyong Xiao
- Department of Radiology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
30
|
Martínez-Rodríguez M, Monteagudo C. CCL27 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1302:113-132. [PMID: 34286445 DOI: 10.1007/978-3-030-62658-7_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chemokines are a group of small proteins which play an important role in leukocyte migration and invasion. They are also involved in the cellular proliferation and migration of tumor cells.Chemokine CCL27 (cutaneous T cell-attracting chemokine, CTACK) is mainly expressed by keratinocytes of the normal epidermis. It is well known that this chemokine plays an important role in several inflammatory diseases of the skin, such as atopic dermatitis, contact dermatitis, and psoriasis. Moreover, several studies have shown an association between CCL27 expression and a variety of neoplasms including skin cancer.In this chapter, we address the role of chemokine CCL27 in the tumor microenvironment in the most relevant cancers of the skin and other anatomical locations. We also make a brief comment on future perspectives and the potential relation of CCL27 with different immunotherapeutic modalities.
Collapse
Affiliation(s)
| | - Carlos Monteagudo
- Department of Pathology, University Clinic Hospital-INCLIVA, University of Valencia, Valencia, Spain.
| |
Collapse
|
31
|
Furue K, Ulzii D, Tanaka Y, Ito T, Tsuji G, Kido‐Nakahara M, Nakahara T, Furue M. Pathogenic implication of epidermal scratch injury in psoriasis and atopic dermatitis. J Dermatol 2020; 47:979-988. [DOI: 10.1111/1346-8138.15507] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Kazuhisa Furue
- Department of Dermatology Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Dugarmaa Ulzii
- Department of Dermatology Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Yuka Tanaka
- Department of Dermatology Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Takamichi Ito
- Department of Dermatology Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Gaku Tsuji
- Department of Dermatology Graduate School of Medical Sciences Kyushu University Fukuoka Japan
- Research and Clinical Center for Yusho and Dioxin Kyushu University Hospital Fukuoka Japan
| | - Makiko Kido‐Nakahara
- Department of Dermatology Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Takeshi Nakahara
- Department of Dermatology Graduate School of Medical Sciences Kyushu University Fukuoka Japan
- Division of Skin Surface Sensing Department of Dermatology Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Masutaka Furue
- Department of Dermatology Graduate School of Medical Sciences Kyushu University Fukuoka Japan
- Research and Clinical Center for Yusho and Dioxin Kyushu University Hospital Fukuoka Japan
- Division of Skin Surface Sensing Department of Dermatology Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| |
Collapse
|
32
|
Cabello-Aguilar S, Alame M, Kon-Sun-Tack F, Fau C, Lacroix M, Colinge J. SingleCellSignalR: inference of intercellular networks from single-cell transcriptomics. Nucleic Acids Res 2020; 48:e55. [PMID: 32196115 PMCID: PMC7261168 DOI: 10.1093/nar/gkaa183] [Citation(s) in RCA: 250] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/23/2020] [Accepted: 03/10/2020] [Indexed: 12/17/2022] Open
Abstract
Single-cell transcriptomics offers unprecedented opportunities to infer the ligand-receptor (LR) interactions underlying cellular networks. We introduce a new, curated LR database and a novel regularized score to perform such inferences. For the first time, we try to assess the confidence in predicted LR interactions and show that our regularized score outperforms other scoring schemes while controlling false positives. SingleCellSignalR is implemented as an open-access R package accessible to entry-level users and available from https://github.com/SCA-IRCM. Analysis results come in a variety of tabular and graphical formats. For instance, we provide a unique network view integrating all the intercellular interactions, and a function relating receptors to expressed intracellular pathways. A detailed comparison of related tools is conducted. Among various examples, we demonstrate SingleCellSignalR on mouse epidermis data and discover an oriented communication structure from external to basal layers.
Collapse
Affiliation(s)
- Simon Cabello-Aguilar
- Institut de Recherche en Cancérologie de Montpellier, Inserm, F-34298 Montpellier, France
- Institut régional du Cancer Montpellier, F-34298 Montpellier, France
- Université de Montpellier, F-34090 Montpellier, France
| | - Mélissa Alame
- Institut de Recherche en Cancérologie de Montpellier, Inserm, F-34298 Montpellier, France
- Institut régional du Cancer Montpellier, F-34298 Montpellier, France
- Université de Montpellier, F-34090 Montpellier, France
- Département d’Hématologie Biologique, CHU Montpellier, Hôpital Saint Eloi, F-34090 Montpellier, France
| | - Fabien Kon-Sun-Tack
- Institut de Recherche en Cancérologie de Montpellier, Inserm, F-34298 Montpellier, France
- Institut régional du Cancer Montpellier, F-34298 Montpellier, France
- Université de Montpellier, F-34090 Montpellier, France
| | - Caroline Fau
- Institut de Recherche en Cancérologie de Montpellier, Inserm, F-34298 Montpellier, France
- Institut régional du Cancer Montpellier, F-34298 Montpellier, France
- Université de Montpellier, F-34090 Montpellier, France
| | - Matthieu Lacroix
- Institut de Recherche en Cancérologie de Montpellier, Inserm, F-34298 Montpellier, France
- Institut régional du Cancer Montpellier, F-34298 Montpellier, France
- Université de Montpellier, F-34090 Montpellier, France
| | - Jacques Colinge
- Institut de Recherche en Cancérologie de Montpellier, Inserm, F-34298 Montpellier, France
- Institut régional du Cancer Montpellier, F-34298 Montpellier, France
- Université de Montpellier, F-34090 Montpellier, France
| |
Collapse
|
33
|
Agarwal S, Sharma A, Bouzeyen R, Deep A, Sharma H, Mangalaparthi KK, Datta KK, Kidwai S, Gowda H, Varadarajan R, Sharma RD, Thakur KG, Singh R. VapBC22 toxin-antitoxin system from Mycobacterium tuberculosis is required for pathogenesis and modulation of host immune response. SCIENCE ADVANCES 2020; 6:eaba6944. [PMID: 32537511 PMCID: PMC7269643 DOI: 10.1126/sciadv.aba6944] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 04/02/2020] [Indexed: 05/16/2023]
Abstract
Virulence-associated protein B and C toxin-antitoxin (TA) systems are widespread in prokaryotes, but their precise role in physiology is poorly understood. We have functionally characterized the VapBC22 TA system from Mycobacterium tuberculosis. Transcriptome analysis revealed that overexpression of VapC22 toxin in M. tuberculosis results in reduced levels of metabolic enzymes and increased levels of ribosomal proteins. Proteomics studies showed reduced expression of virulence-associated proteins and increased levels of cognate antitoxin, VapB22 in the ΔvapC22 mutant strain. Furthermore, both the ΔvapC22 mutant and VapB22 overexpression strains of M. tuberculosis were susceptible to killing upon exposure to oxidative stress and showed attenuated growth in guinea pigs and mice. Host transcriptome analysis suggests upregulation of the transcripts involved in innate immune responses and tissue remodeling in mice infected with the ΔvapC22 mutant strain. Together, we demonstrate that the VapBC22 TA system belongs to a key regulatory network and is essential for M. tuberculosis pathogenesis.
Collapse
Affiliation(s)
- Sakshi Agarwal
- Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Harya na-121001, India
| | - Arun Sharma
- Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Harya na-121001, India
| | - Rania Bouzeyen
- Institut Pasteur de Tunis, LTCII, LR11IPT02, Tunis 1002, Tunisia
| | - Amar Deep
- Structural Biology Laboratory, Council of Scientific and Industrial Research–Institute of Microbial Technology, Chandigarh 160036, India
| | - Harsh Sharma
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurugr am-122413, India
| | | | | | - Saqib Kidwai
- Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Harya na-121001, India
| | - Harsha Gowda
- Institute of Bioinformatics, Bangalore 560066, India
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | | | - Ravi Datta Sharma
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurugr am-122413, India
| | - Krishan Gopal Thakur
- Structural Biology Laboratory, Council of Scientific and Industrial Research–Institute of Microbial Technology, Chandigarh 160036, India
| | - Ramandeep Singh
- Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Harya na-121001, India
- Corresponding author.
| |
Collapse
|
34
|
Chen Z, Haus JM, Chen L, Wu SC, Urao N, Koh TJ, Minshall RD. CCL28-induced CCR10/eNOS interaction in angiogenesis and skin wound healing. FASEB J 2020; 34:5838-5850. [PMID: 32124475 DOI: 10.1096/fj.201902060r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 02/06/2020] [Accepted: 02/20/2020] [Indexed: 12/25/2022]
Abstract
Chemokines and their receptors play important roles in vascular homeostasis, development, and angiogenesis. Little is known regarding the molecular signaling mechanisms activated by CCL28 chemokine via its primary receptor CCR10 in endothelial cells (ECs). Here, we test the hypothesis that CCL28/CCR10 signaling plays an important role in regulating skin wound angiogenesis through endothelial nitric oxide synthase (eNOS)-dependent Src, PI3K, and MAPK signaling. We observed nitric oxide (NO) production in human primary ECs stimulated with exogenous CCL28, which also induced direct binding of CCR10 and eNOS resulting in inhibition of eNOS activity. Knockdown of CCR10 with siRNA lead to reduced eNOS expression and tube formation suggesting the involvement of CCR10 in EC angiogenesis. Based on this interaction, we engineered a myristoylated 7 amino acid CCR10-binding domain (Myr-CBD7) peptide and showed that this can block eNOS interaction with CCR10, but not with calmodulin, resulting in upregulation of eNOS activity. Importantly, topical administration of Myr-CBD7 peptide on mouse dermal wounds not only blocked CCR10-eNOS interaction, but also enhanced expression of eNOS, CD31, and IL-4 with reduction of CCL28 and IL-6 levels associated with improved wound healing. These results point to a potential therapeutic strategy to upregulate NO bioavailability, enhance angiogenesis, and improve wound healing by disrupting CCL28-activated CCR10-eNOS interaction.
Collapse
Affiliation(s)
- Zhenlong Chen
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Jacob M Haus
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Lin Chen
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL, USA.,Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, USA
| | - Stephanie C Wu
- Center for Lower Extremity Ambulatory Research (CLEAR), Dr. William M. Scholl College of Podiatric Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Norifumi Urao
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA.,Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, USA
| | - Timothy J Koh
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA.,Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, USA
| | - Richard D Minshall
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
35
|
Chen YT, Hsu H, Lin CC, Pan SY, Liu SY, Wu CF, Tsai PZ, Liao CT, Cheng HT, Chiang WC, Chen YM, Chu TS, Lin SL. Inflammatory macrophages switch to CCL17-expressing phenotype and promote peritoneal fibrosis. J Pathol 2019; 250:55-66. [PMID: 31579932 DOI: 10.1002/path.5350] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 09/10/2019] [Accepted: 09/19/2019] [Indexed: 12/30/2022]
Abstract
Peritoneal fibrosis remains a problem in kidney failure patients treated with peritoneal dialysis. Severe peritoneal fibrosis with encapsulation or encapsulating peritoneal sclerosis is devastating and life-threatening. Although submesothelial fibroblasts as the major precursor of scar-producing myofibroblasts in animal models and M2 macrophage (Mϕ)-derived chemokines in peritoneal effluents of patients before diagnosis of encapsulating peritoneal sclerosis have been identified, attenuation of peritoneal fibrosis is an unmet medical need partly because the mechanism for cross talk between Mϕs and fibroblasts remains unclear. We use a sodium hypochlorite-induced mouse model akin to clinical encapsulated peritoneal sclerosis to study how the peritoneal Mϕs activate fibroblasts and fibrosis. Sodium hypochlorite induces the disappearance of CD11bhigh F4/80high resident Mϕs but accumulation of CD11bint F4/80int inflammatory Mϕs (InfMϕs) through recruiting blood monocytes and activating local cell proliferation. InfMϕs switch to express chemokine (C-C motif) ligand 17 (CCL17), CCL22, and arginase-1 from day 2 after hypochlorite injury. More than 75% of InfMϕs undergo genetic recombination by Csf1r-driven Cre recombinase, providing the possibility to reduce myofibroblasts and fibrosis by diphtheria toxin-induced Mϕ ablation from day 2 after injury. Furthermore, administration of antibody against CCL17 can reduce Mϕs, myofibroblasts, fibrosis, and improve peritoneal function after injury. Mechanistically, CCL17 stimulates migration and collagen production of submesothelial fibroblasts in culture. By breeding mice that are induced to express red fluorescent protein in Mϕs and green fluorescence protein (GFP) in Col1a1-expressing cells, we confirmed that Mϕs do not produce collagen in peritoneum before and after injury. However, small numbers of fibrocytes are found in fibrotic peritoneum of chimeric mice with bone marrow from Col1a1-GFP reporter mice, but they do not contribute to myofibroblasts. These data demonstrate that InfMϕs switch to pro-fibrotic phenotype and activate peritoneal fibroblasts through CCL17 after injury. CCL17 blockade in patients with peritoneal fibrosis may provide a novel therapy. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yi-Ting Chen
- Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei, Taiwan.,Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Internal Medicine, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Hao Hsu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chi-Chun Lin
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Szu-Yu Pan
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Renal Division, Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Shin-Yun Liu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ching-Fang Wu
- Department of Internal Medicine, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Pei-Zhen Tsai
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Te Liao
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Hui-Teng Cheng
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu, Taiwan
| | - Wen-Chih Chiang
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yung-Ming Chen
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Tzong-Shinn Chu
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shuei-Liong Lin
- Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei, Taiwan.,Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
36
|
Tissue Iron Promotes Wound Repair via M2 Macrophage Polarization and the Chemokine (C-C Motif) Ligands 17 and 22. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:2196-2208. [PMID: 31465751 DOI: 10.1016/j.ajpath.2019.07.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/16/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022]
Abstract
Macrophages are important for effective iron recycling and erythropoiesis, but they also play a crucial role in wound healing, orchestrating tissue repair. Recently, we demonstrated a significant accumulation of iron in healing wounds and a requirement of iron for effective repair. Herein, we sought to determine the influence of iron on macrophage function in the context of wound healing. Interestingly, wound macrophages extensively sequestered iron throughout healing, associated with a prohealing M2 phenotype. In delayed healing diabetic mouse wounds, both macrophage polarization and iron sequestration were impaired. In vitro studies revealed that iron promotes differentiation, while skewing macrophages toward a hypersecretory M2-like polarization state. These macrophages produced high levels of chemokine (C-C motif) ligands 17 and 22, promoting wound reepithelialization and extracellular matrix deposition in a human ex vivo wound healing model. Together, these findings reveal a novel, unappreciated role for iron in modulating macrophage behavior to promote subsequent wound repair. These findings support therapeutic evaluation of iron use to promote wound healing in the clinic.
Collapse
|
37
|
Kobayashi S, Sakurai T, So T, Shiota Y, Asao A, Phung HT, Tanaka R, Kawabe T, Maruyama T, Kanno E, Kawakami K, Owada Y, Ishii N. TNF Receptor-Associated Factor 5 Limits Function of Plasmacytoid Dendritic Cells by Controlling IFN Regulatory Factor 5 Expression. THE JOURNAL OF IMMUNOLOGY 2019; 203:1447-1456. [PMID: 31420465 DOI: 10.4049/jimmunol.1900188] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/19/2019] [Indexed: 12/15/2022]
Abstract
The physiological functions of TNF receptor-associated factor 5 (TRAF5) in the skin inflammation and wound healing process are not well characterized. We found that Traf5 -/- mice exhibited an accelerated skin wound healing as compared with wild-type counterparts. The augmented wound closure in Traf5 -/- mice was associated with a massive accumulation of plasmacytoid dendritic cells (pDCs) into skin wounds and an enhanced expression of genes related to wound repair at skin sites. In accordance with this result, adoptive transfer of Traf5 -/- pDCs, but not wild-type pDCs, into the injured skin area in wild-type recipient mice significantly promoted skin wound healing. The expression of skin-tropic chemokine receptor CXCR3 was significantly upregulated in Traf5-/- pDCs, and treatment with a CXCR3 inhibitor cancelled the promoted wound healing in Traf5-/- mice, suggesting a pivotal role of CXCR3 in pDC-dependent wound healing. Traf5 -/- pDCs displayed significantly higher expression of IFN regulatory factor 5 (IRF5), which correlated with greater induction of proinflammatory cytokine genes and CXCR3 protein after stimulation with TLR ligands. Consistently, transduction of exogeneous TRAF5 in Traf5-/- pDCs normalized the levels of abnormally elevated proinflammatory molecules, including IRF5 and CXCR3. Furthermore, knockdown of IRF5 also rescued the abnormal phenotypes of Traf5-/- pDCs. Therefore, the higher expression and induction of IRF5 in Traf5-/- pDCs causes proinflammatory and skin-tropic characteristics of the pDCs, which may accelerate skin wound healing responses. Collectively, our results uncover a novel role of TRAF5 in skin wound healing that is mediated by IRF5-dependent function of pDCs.
Collapse
Affiliation(s)
- Shuhei Kobayashi
- Department of Microbiology and Immunology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Tsuyoshi Sakurai
- Department of Microbiology and Immunology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Takanori So
- Department of Microbiology and Immunology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan.,Laboratory of Molecular Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Yuka Shiota
- Department of Microbiology and Immunology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Atsuko Asao
- Department of Microbiology and Immunology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Hai The Phung
- Department of Microbiology and Immunology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Riou Tanaka
- Department of Microbiology and Immunology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Takeshi Kawabe
- Department of Microbiology and Immunology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Takashi Maruyama
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, Akita University, Akita 010-8543, Japan
| | - Emi Kanno
- Department of Science of Nursing Practice, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Kazuyoshi Kawakami
- Department of Medical Microbiology, Mycology and Immunology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan; and
| | - Yuji Owada
- Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Naoto Ishii
- Department of Microbiology and Immunology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan;
| |
Collapse
|
38
|
Papi M, Papi C. Biologics in Microangiopathic Wounds. INT J LOW EXTR WOUND 2018. [DOI: 10.1177/1534734618813767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the last decades the possibility to diagnose a skin ulcer has greatly improved. We learnt that a consistent percentage of nonhealing ulcers may be caused by a microangiopathic disorder that has not been properly investigated and cured. Pathogenetically, we can distinguish 2 main groups: (1) ulcers due to inflammatory microangiopathy, mainly including cutaneous small and medium vessel vasculitis, pyoderma gangrenosum, and connective tissue diseases, and (2) ulcers due to occlusive microangiopathy. The group of microangiopathic occlusive ulcers is more heterogeneous and includes different disorders ranging from livedo vasculopathy to calciphylaxis, hydroxyurea-induced ulcers, antiphospholipid antibodies ulcers, and various other types. These conditions can induce thromboses or anatomo-functional occlusion of cutaneous microvessels. Despite different physiopathologic mechanisms, the ulcer resulting from a primitive microangiopathy may receive basic treatments that are in the complex similar to other pathogenetically different wounds, including MOIST-based local therapy and elastic compression when it is not contraindicated. Persistent inflammatory processes are increasingly demonstrated as responsible for the chronicity of many skin ulcers. New data concerning the biological phases of wound healing and the molecules that play crucial roles in this process suggested the use of new specific therapies. Some of them such as growth factors and platelet-rich plasma are prevalently used as topical biologic agents with variable benefits. In recent years, a new class of systemic anti-inflammatory molecules, better known as biologic drugs, have been introduced in the cure of chronic inflammatory diseases that can induce microangiopathic injuries and ulcerative complication. They enlarged the therapeutic options in case of nonresponder microangiopathic ulcers and could represent a future model of “pathogenetically based” therapy of skin ulcers.
Collapse
Affiliation(s)
- Massimo Papi
- National Institute of Health, Migration and Poverty, Rome, Italy
| | - Claudia Papi
- Catholic University Policlinico A. Gemelli, Rome, Italy
| |
Collapse
|
39
|
Jacquelot N, Duong CPM, Belz GT, Zitvogel L. Targeting Chemokines and Chemokine Receptors in Melanoma and Other Cancers. Front Immunol 2018; 9:2480. [PMID: 30420855 PMCID: PMC6215820 DOI: 10.3389/fimmu.2018.02480] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/08/2018] [Indexed: 12/19/2022] Open
Abstract
The tumor microenvironment is highly heterogeneous. It is composed of a diverse array of immune cells that are recruited continuously into lesions. They are guided into the tumor through interactions between chemokines and their receptors. A variety of chemokine receptors are expressed on the surface of both tumor and immune cells rendering them sensitive to multiple stimuli that can subsequently influence their migration and function. These features significantly impact tumor fate and are critical in melanoma control and progression. Indeed, particular chemokine receptors expressed on tumor and immune cells are strongly associated with patient prognosis. Thus, potential targeting of chemokine receptors is highly attractive as a means to quench or eliminate unconstrained tumor cell growth.
Collapse
Affiliation(s)
- Nicolas Jacquelot
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Connie P M Duong
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM U1015, Villejuif, France
| | - Gabrielle T Belz
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM U1015, Villejuif, France.,Faculty of Medicine, Paris Sud/Paris XI University, LeKremlin-Bicêtre, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| |
Collapse
|