1
|
Shahin H, Belcastro L, Das J, Perdiki Grigoriadi M, Saager RB, Steinvall I, Sjöberg F, Olofsson P, Elmasry M, El-Serafi AT. MicroRNA-155 mediates multiple gene regulations pertinent to the role of human adipose-derived mesenchymal stem cells in skin regeneration. Front Bioeng Biotechnol 2024; 12:1328504. [PMID: 38562669 PMCID: PMC10982420 DOI: 10.3389/fbioe.2024.1328504] [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: 10/26/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction: The role of Adipose-derived mesenchymal stem cells (AD-MSCs) in skin wound healing remains to be fully characterized. This study aims to evaluate the regenerative potential of autologous AD-MSCs in a non-healing porcine wound model, in addition to elucidate key miRNA-mediated epigenetic regulations that underlie the regenerative potential of AD-MSCs in wounds. Methods: The regenerative potential of autologous AD-MSCs was evaluated in porcine model using histopathology and spatial frequency domain imaging. Then, the correlations between miRNAs and proteins of AD-MSCs were evaluated using an integration analysis in primary human AD-MSCs in comparison to primary human keratinocytes. Transfection study of AD-MSCs was conducted to validate the bioinformatics data. Results: Autologous porcine AD-MSCs improved wound epithelialization and skin properties in comparison to control wounds. We identified 26 proteins upregulated in human AD-MSCs, including growth and angiogenic factors, chemokines and inflammatory cytokines. Pathway enrichment analysis highlighted cell signalling-associated pathways and immunomodulatory pathways. miRNA-target modelling revealed regulations related to genes encoding for 16 upregulated proteins. miR-155-5p was predicted to regulate Fibroblast growth factor 2 and 7, C-C motif chemokine ligand 2 and Vascular cell adhesion molecule 1. Transfecting human AD-MSCs cell line with anti-miR-155 showed transient gene silencing of the four proteins at 24 h post-transfection. Discussion: This study proposes a positive miR-155-mediated gene regulation of key factors involved in wound healing. The study represents a promising approach for miRNA-based and cell-free regenerative treatment for difficult-to-heal wounds. The therapeutic potential of miR-155 and its identified targets should be further explored in-vivo.
Collapse
Affiliation(s)
- Hady Shahin
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linkoping University, Linköping, Sweden
- Faculty of Biotechnology, Modern Sciences and Arts University, October City, Cairo, Egypt
| | - Luigi Belcastro
- Department of Biomedical Engineering, Linkoping University, Linköping, Sweden
| | - Jyotirmoy Das
- Bioinformatics Unit, Core Facility (KEF), Faculty of Medicine and Health Sciences (BKV), Linköping University, Linköping, Sweden
- Clinical Genomics Linköping, SciLife Laboratory, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | | | - Rolf B. Saager
- Department of Biomedical Engineering, Linkoping University, Linköping, Sweden
| | - Ingrid Steinvall
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
| | - Folke Sjöberg
- Department of Biomedical and Clinical Sciences, Linkoping University, Linköping, Sweden
| | - Pia Olofsson
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
| | - Moustafa Elmasry
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
| | - Ahmed T. El-Serafi
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linkoping University, Linköping, Sweden
| |
Collapse
|
2
|
Zlobina K, Malekos E, Chen H, Gomez M. Robust classification of wound healing stages in both mice and humans for acute and burn wounds based on transcriptomic data. BMC Bioinformatics 2023; 24:166. [PMID: 37098473 PMCID: PMC10127407 DOI: 10.1186/s12859-023-05295-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 04/17/2023] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND Wound healing involves careful coordination among various cell types carrying out unique or even multifaceted functions. The abstraction of this complex dynamic process into four primary wound stages is essential to the study of wound care for timing treatment and tracking wound progression. For example, a treatment that may promote healing in the inflammatory stage may prove detrimental in the proliferative stage. Additionally, the time scale of individual responses varies widely across and within the same species. Therefore, a robust method to assess wound stages can help advance translational work from animals to humans. RESULTS In this work, we present a data-driven model that robustly identifies the dominant wound healing stage using transcriptomic data from biopsies gathered from mouse and human wounds, both burn and surgical. A training dataset composed of publicly available transcriptomic arrays is used to derive 58 shared genes that are commonly differentially expressed. They are divided into 5 clusters based on temporal gene expression dynamics. The clusters represent a 5-dimensional parametric space containing the wound healing trajectory. We then create a mathematical classification algorithm in the 5-dimensional space and demonstrate that it can distinguish between the four stages of wound healing: hemostasis, inflammation, proliferation, and remodeling. CONCLUSIONS In this work, we present an algorithm for wound stage detection based on gene expression. This work suggests that there are universal characteristics of gene expression in wound healing stages despite the seeming disparities across species and wounds. Our algorithm performs well for human and mouse wounds of both burn and surgical types. The algorithm has the potential to serve as a diagnostic tool that can advance precision wound care by providing a way of tracking wound healing progression with more accuracy and finer temporal resolution compared to visual indicators. This increases the potential for preventive action.
Collapse
Affiliation(s)
| | | | - Han Chen
- University of California, Santa Cruz, CA, USA
| | | |
Collapse
|
3
|
Zhang Y, Guo C, Yang S, Elkharti M, Liu R, Sun MZ, Liu S. NONHSAT021545/miR-330-3p/EREG: A Cooperative Axis in Breast Cancer Prognosis and Treatment. J Clin Med 2023; 12:jcm12072478. [PMID: 37048561 PMCID: PMC10094950 DOI: 10.3390/jcm12072478] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/28/2023] [Accepted: 03/13/2023] [Indexed: 04/14/2023] Open
Abstract
Lymphatic metastasis is the most common form in breast cancer (BC) progression. Previously, we observed that lnc045874, a most conservative homology of Homo Sapiens NONHSAT021545 (lnc021545), miR-330-3p, and EREG may have some effects in mouse hepatocarcinoma cell lines with different lymphatic metastasis potentials. Through data from TCGA and GEO database analysis, we speculated that miR-330-3p might be a tumor promoter, while EREG could be a tumor suppressor in BC. MiR-330-3p was upregulated, while lnc021545 and EREG were downregulated in 50 BC tissues. MiR-330-3p advanced the metastatic behaviors of BC cells, whereas lnc021545 and EREG resulted in the opposite effects. The three molecules' expressions were correlated respectively and showed that miR-330-3p targeted lnc021545 and EREG to affect their expressions. Lnc021545/miR-330-3p axis affected BC metastasis by regulating EREG in epithelial-to-mesenchymal transition. In 50 BC patients, these three molecules and their cooperation are associated with aggressive tumor phenotypes, patient outcomes, and trastuzumab therapy. We finally discovered that lnc021545, miR-330-3p, and EREG formed a multi-gene co-regulation system that affected the metastasis of BC and the cooperation reflects the synergistic effects of the three molecules, recommending that their cooperation may provide a more accurate index for anti-metastasis therapeutic and prognostic evaluation of BC.
Collapse
Affiliation(s)
- Yunkun Zhang
- Department of Biochemistry, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
- Department of Pathology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Chunmei Guo
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Siwen Yang
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Maroua Elkharti
- Department of Biochemistry, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Rui Liu
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Ming-Zhong Sun
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Shuqing Liu
- Department of Biochemistry, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
4
|
Akbarian M, Bertassoni LE, Tayebi L. Biological aspects in controlling angiogenesis: current progress. Cell Mol Life Sci 2022; 79:349. [PMID: 35672585 PMCID: PMC10171722 DOI: 10.1007/s00018-022-04348-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/01/2022] [Accepted: 05/03/2022] [Indexed: 12/25/2022]
Abstract
All living beings continue their life by receiving energy and by excreting waste products. In animals, the arteries are the pathways of these transfers to the cells. Angiogenesis, the formation of the arteries by the development of pre-existed parental blood vessels, is a phenomenon that occurs naturally during puberty due to certain physiological processes such as menstruation, wound healing, or the adaptation of athletes' bodies during exercise. Nonetheless, the same life-giving process also occurs frequently in some patients and, conversely, occurs slowly in some physiological problems, such as cancer and diabetes, so inhibiting angiogenesis has been considered to be one of the important strategies to fight these diseases. Accordingly, in tissue engineering and regenerative medicine, the highly controlled process of angiogenesis is very important in tissue repairing. Excessive angiogenesis can promote tumor progression and lack of enough angiogensis can hinder tissue repair. Thereby, both excessive and deficient angiogenesis can be problematic, this review article introduces and describes the types of factors involved in controlling angiogenesis. Considering all of the existing strategies, we will try to lay out the latest knowledge that deals with stimulating/inhibiting the angiogenesis. At the end of the article, owing to the early-reviewed mechanical aspects that overshadow angiogenesis, the strategies of angiogenesis in tissue engineering will be discussed.
Collapse
Affiliation(s)
- Mohsen Akbarian
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Luiz E Bertassoni
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA.
| |
Collapse
|
5
|
Cheng WL, Feng PH, Lee KY, Chen KY, Sun WL, Van Hiep N, Luo CS, Wu SM. The Role of EREG/EGFR Pathway in Tumor Progression. Int J Mol Sci 2021; 22:ijms222312828. [PMID: 34884633 PMCID: PMC8657471 DOI: 10.3390/ijms222312828] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Aberrant activation of the epidermal growth factor receptor (EGFR/ERBB1) by erythroblastic leukemia viral oncogene homolog (ERBB) ligands contributes to various tumor malignancies, including lung cancer and colorectal cancer (CRC). Epiregulin (EREG) is one of the EGFR ligands and is low expressed in most normal tissues. Elevated EREG in various cancers mainly activates EGFR signaling pathways and promotes cancer progression. Notably, a higher EREG expression level in CRC with wild-type Kirsten rat sarcoma viral oncogene homolog (KRAS) is related to better efficacy of therapeutic treatment. By contrast, the resistance of anti-EGFR therapy in CRC was driven by low EREG expression, aberrant genetic mutation and signal pathway alterations. Additionally, EREG overexpression in non-small cell lung cancer (NSCLC) is anticipated to be a therapeutic target for EGFR-tyrosine kinase inhibitor (EGFR-TKI). However, recent findings indicate that EREG derived from macrophages promotes NSCLC cell resistance to EGFR-TKI treatment. The emerging events of EREG-mediated tumor promotion signals are generated by autocrine and paracrine loops that arise from tumor epithelial cells, fibroblasts, and macrophages in the tumor microenvironment (TME). The TME is a crucial element for the development of various cancer types and drug resistance. The regulation of EREG/EGFR pathways depends on distinct oncogenic driver mutations and cell contexts that allows specific pharmacological targeting alone or combinational treatment for tailored therapy. Novel strategies targeting EREG/EGFR, tumor-associated macrophages, and alternative activation oncoproteins are under development or undergoing clinical trials. In this review, we summarize the clinical outcomes of EREG expression and the interaction of this ligand in the TME. The EREG/EGFR pathway may be a potential target and may be combined with other driver mutation targets to combat specific cancers.
Collapse
Affiliation(s)
- Wan-Li Cheng
- Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan;
- Division of Cardiovascular Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (P.-H.F.); (K.-Y.L.); (K.-Y.C.); (W.-L.S.); (N.V.H.); (C.-S.L.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (P.-H.F.); (K.-Y.L.); (K.-Y.C.); (W.-L.S.); (N.V.H.); (C.-S.L.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (P.-H.F.); (K.-Y.L.); (K.-Y.C.); (W.-L.S.); (N.V.H.); (C.-S.L.)
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Lun Sun
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (P.-H.F.); (K.-Y.L.); (K.-Y.C.); (W.-L.S.); (N.V.H.); (C.-S.L.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Nguyen Van Hiep
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (P.-H.F.); (K.-Y.L.); (K.-Y.C.); (W.-L.S.); (N.V.H.); (C.-S.L.)
- International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Ching-Shan Luo
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (P.-H.F.); (K.-Y.L.); (K.-Y.C.); (W.-L.S.); (N.V.H.); (C.-S.L.)
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (P.-H.F.); (K.-Y.L.); (K.-Y.C.); (W.-L.S.); (N.V.H.); (C.-S.L.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence:
| |
Collapse
|
6
|
Oropallo AR, Serena TE, Armstrong DG, Niederauer MQ. Molecular Biomarkers of Oxygen Therapy in Patients with Diabetic Foot Ulcers. Biomolecules 2021; 11:biom11070925. [PMID: 34206433 PMCID: PMC8301753 DOI: 10.3390/biom11070925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/13/2021] [Accepted: 06/19/2021] [Indexed: 12/28/2022] Open
Abstract
Hyperbaric oxygen therapy (HBOT) and topical oxygen therapy (TOT) including continuous diffuse oxygen therapy (CDOT) are often utilized to enhance wound healing in patients with diabetic foot ulcerations. High pressure pure oxygen assists in the oxygenation of hypoxic wounds to increase perfusion. Although oxygen therapy provides wound healing benefits to some patients with diabetic foot ulcers, it is currently performed from clinical examination and imaging. Data suggest that oxygen therapy promotes wound healing via angiogenesis, the creation of new blood vessels. Molecular biomarkers relating to tissue inflammation, repair, and healing have been identified. Predictive biomarkers can be used to identify patients who will most likely benefit from this specialized treatment. In diabetic foot ulcerations, specifically, certain biomarkers have been linked to factors involving angiogenesis and inflammation, two crucial aspects of wound healing. In this review, the mechanism of how oxygen works in wound healing on a physiological basis, such as cell metabolism and growth factor signaling transduction is detailed. Additionally, observable clinical outcomes such as collagen formation, angiogenesis, respiratory burst and cell proliferation are described. The scientific evidence for the impact of oxygen on biomolecular pathways and its relationship to the outcomes in clinical research is discussed in this narrative review.
Collapse
Affiliation(s)
- Alisha R. Oropallo
- Comprehensive Wound Healing Center and Hyperbarics, Department of Vascular Surgery, Zucker School of Medicine Hofstra/Northwell, Hempstead, NY 11549, USA
- Correspondence: ; Tel.: +1-516-233-3780
| | | | - David G. Armstrong
- Limb Preservation Program, Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA;
| | | |
Collapse
|
7
|
Iwata H, Haga N, Ujiie H. Possible role of epiregulin from dermal fibroblasts in the keratinocyte hyperproliferation of psoriasis. J Dermatol 2021; 48:1433-1438. [PMID: 34128258 DOI: 10.1111/1346-8138.16003] [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: 03/16/2021] [Revised: 04/26/2021] [Accepted: 05/09/2021] [Indexed: 11/30/2022]
Abstract
Psoriasis, an immune-mediated inflammatory disease, is characterized by keratinocyte hyperproliferation. Tumor necrosis factor (TNF)-α, interleukin (IL)-23, and IL-17A play critical roles in the pathogenesis of psoriasis. IL-17A secreted by T-helper 17 acts more directly against keratinocytes than TNF-α or IL-23 do. Regarding the receptors of cytokines, fibroblasts also express receptors against IL-17A and TNF-α, and induce the production of growth factors. Epiregulin (EREG), an epidermal growth factor receptor ligand, is produced by both keratinocytes and fibroblasts. EREG enhances keratinocyte proliferation and differentiation. We hypothesized that fibroblasts stimulated with IL-17A and/or TNF-α may play a role in epidermal hyperproliferation through the production of epidermal growth factors in psoriasis. The mRNA expression of EREG was found to be significantly upregulated by co-stimulation with IL-17A and TNF-α (mean, 49.2-fold). Furthermore, the stimulation with TNF-α alone, but not IL-17A alone, induced significant increases. Immunofluorescent staining demonstrated that the protein expression level of EREG was also increased in fibroblasts stimulated with these cytokines. Stimulation with EREG significantly enhanced keratinocyte proliferation in vitro. In human psoriatic patients' skin, immunofluorescence staining of EREG showed significantly high intensity in the dermis of lesional skin. In conclusion, cytokine stimulation with TNF-α and IL-17A induces the overexpression of EREG from dermal fibroblasts in the lesional skin of psoriasis, and plays a role in epidermal hyperproliferation.
Collapse
Affiliation(s)
- Hiroaki Iwata
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Naoya Haga
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hideyuki Ujiie
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
8
|
Urolithin A augments angiogenic pathways in skeletal muscle by bolstering NAD + and SIRT1. Sci Rep 2020; 10:20184. [PMID: 33214614 PMCID: PMC7678835 DOI: 10.1038/s41598-020-76564-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/12/2020] [Indexed: 12/27/2022] Open
Abstract
Urolithin A (UA) is a natural compound that is known to improve muscle function. In this work we sought to evaluate the effect of UA on muscle angiogenesis and identify the underlying molecular mechanisms. C57BL/6 mice were administered with UA (10 mg/body weight) for 12–16 weeks. ATP levels and NAD+ levels were measured using in vivo 31P NMR and HPLC, respectively. UA significantly increased ATP and NAD+ levels in mice skeletal muscle. Unbiased transcriptomics analysis followed by Ingenuity Pathway Analysis (IPA) revealed upregulation of angiogenic pathways upon UA supplementation in murine muscle. The expression of the differentially regulated genes were validated using quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC). Angiogenic markers such as VEGFA and CDH5 which were blunted in skeletal muscles of 28 week old mice were found to be upregulated upon UA supplementation. Such augmentation of skeletal muscle vascularization was found to be bolstered via Silent information regulator 1 (SIRT1) and peroxisome proliferator-activated receptor-gamma coactivator-1-alpha (PGC-1α) pathway. Inhibition of SIRT1 by selisistat EX527 blunted UA-induced angiogenic markers in C2C12 cells. Thus this work provides maiden evidence demonstrating that UA supplementation bolsters skeletal muscle ATP and NAD+ levels causing upregulated angiogenic pathways via a SIRT1-PGC-1α pathway.
Collapse
|
9
|
Time course analysis of large-scale gene expression in incised muscle using correspondence analysis. PLoS One 2020; 15:e0230737. [PMID: 32210454 PMCID: PMC7094855 DOI: 10.1371/journal.pone.0230737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/06/2020] [Indexed: 11/23/2022] Open
Abstract
Studying the time course of gene expression in injured skeletal muscle would help to estimate the timing of injuries. In this study, we investigated large-scale gene expression in incision-injured mouse skeletal muscle by DNA microarray using correspondence analysis (CA). Biceps femoris muscle samples were collected 6, 12, and 24 hours after injury, and RNA was extracted and prepared for microarray analysis. On a 2-dimensional plot by CA, the genes (row score coordinate) located farther from each time series (column score coordinate) had more upregulation at particular times. Each gene was situated in 6 subdivided triangular areas according to the magnitude of the relationship of the fold change (FC) value at each time point compared to the control. In each area, genes for which the ratios of two particular FC values were close to 1 were distributed along the two border lines. There was a tendency for genes whose FC values were almost equal to be distributed near the intersection of these 6 areas. Therefore, the gene marker candidates for estimation of the timing of injuries were detectable according to the location on the CA plot. Moreover, gene sets created by a specific gene and its surrounding genes were composed of genes that showed similar or identical fluctuation patterns to the specific gene. In various analyses on these sets, significant gene ontology term and pathway activity may reflect changes in specific genes. In conclusion, analyses of gene sets based on CA plots is effective for investigation of the time-dependent fluctuation in gene expression after injury.
Collapse
|
10
|
Knockout of MicroRNA-155 Ameliorates the Th17/Th9 Immune Response and Promotes Wound Healing. Curr Med Sci 2019; 39:954-964. [PMID: 31845227 DOI: 10.1007/s11596-019-2128-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 10/20/2019] [Indexed: 01/15/2023]
Abstract
MiRNAs are integral for maintaining immune homeostasis and self-tolerance. In this study, qPCR analyses were performed to determine which miRNAs play an important role in wound healing. Next, an experiment in a model of wound healing was performed, and histology, mRNA expression and T-cell subpopulations in wound tissue were analyzed. The accelerated experiments were performed by local injection of either rIL-17A and/or rIL-9 after wound healing. In vitro, the differentiation of Th17/Th9 in miR-155+/+ or miR-155-/- mice was investigated, and the target genes of miR155 were analyzed. From our findings, miR-155-/- in mice promoted wound healing and weakened T cell-mediated inflammation, especially in IL-17/IL-9, and less severe skin fibrosis developed in the mice. rIL-17A and/or rIL-9 could exacerbate inflammatory injury and delay wound healing. We also demonstrated that miR-155-/- led to a defect in the differentiation of Th17/Th9 in vitro, and this effect of IL-17/IL-9 might be related to the expression of C-maf, which is a target gene of miR155. MiR-155 regulated IL-17/IL-9-related inflammation in wound healing and might be a potential therapeutic target to attenuate the inflammatory response in wound tissue and promote the closure of wound injuries.
Collapse
|
11
|
Das A, Masry MSE, Gnyawali SC, Ghatak S, Singh K, Stewart R, Lewis M, Saha A, Gordillo G, Khanna S. Skin Transcriptome of Middle-Aged Women Supplemented With Natural Herbo-mineral Shilajit Shows Induction of Microvascular and Extracellular Matrix Mechanisms. J Am Coll Nutr 2019; 38:526-536. [PMID: 31161927 PMCID: PMC7027386 DOI: 10.1080/07315724.2018.1564088] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/21/2018] [Accepted: 12/23/2018] [Indexed: 12/20/2022]
Abstract
Objective: Shilajit is a pale-brown to blackish-brown organic mineral substance available from Himalayan rocks. We demonstrated that in type I obese humans, shilajit supplementation significantly upregulated extracellular matrix (ECM)-related genes in the skeletal muscle. Such an effect was highly synergistic with exercise. The present study (clinicaltrials.gov NCT02762032) aimed to evaluate the effects of shilajit supplementation on skin gene expression profile and microperfusion in healthy adult females. Methods: The study design comprised six total study visits including a baseline visit (V1) and a final 14-week visit (V6) following oral shilajit supplementation (125 or 250 mg bid). A skin biopsy of the left inner upper arm of each subject was collected at visit 2 and visit 6 for gene expression profiling using Affymetrix Clariom™ D Assay. Skin perfusion was determined by MATLAB processing of dermascopic images. Transcriptome data were normalized and subjected to statistical analysis. The differentially regulated genes were subjected to Ingenuity Pathway Analysis (IPA®). The expression of the differentially regulated genes identified by IPA® were verified using real-time polymerase chain reaction (RT-PCR). Results: Supplementation with shilajit for 14 weeks was not associated with any reported adverse effect within this period. At a higher dose (250 mg bid), shilajit improved skin perfusion when compared to baseline or the placebo. Pathway analysis identified shilajit-inducible genes relevant to endothelial cell migration, growth of blood vessels, and ECM which were validated by quantitative real-time polymerase chain reaction (RT-PCR) analysis. Conclusions: This work provides maiden evidence demonstrating that oral shilajit supplementation in adult healthy women induced genes relevant to endothelial cell migration and growth of blood vessels. Shilajit supplementation improved skin microperfusion.
Collapse
Affiliation(s)
- Amitava Das
- Department of Surgery, Indiana Center for Regenerative
Medicine and Engineering, Indiana University School of Medicine, Indianapolis,
IN
- Department of Surgery, The Ohio State University, Wexner
Medical Center, Columbus, Ohio
| | - Mohamed S. El Masry
- Department of Surgery, Indiana Center for Regenerative
Medicine and Engineering, Indiana University School of Medicine, Indianapolis,
IN
- Department of Surgery, The Ohio State University, Wexner
Medical Center, Columbus, Ohio
- Department of Plastic and Reconstructive Surgery, Zagazig
University, Zagazig, Egypt
| | - Surya C. Gnyawali
- Department of Surgery, The Ohio State University, Wexner
Medical Center, Columbus, Ohio
| | - Subhadip Ghatak
- Department of Surgery, The Ohio State University, Wexner
Medical Center, Columbus, Ohio
- Department of Plastic Surgery, Indiana University School of
Medicine, Indianapolis, IN
| | - Kanhaiya Singh
- Department of Surgery, Indiana Center for Regenerative
Medicine and Engineering, Indiana University School of Medicine, Indianapolis,
IN
- Department of Surgery, The Ohio State University, Wexner
Medical Center, Columbus, Ohio
| | - Richard Stewart
- Department of Surgery, The Ohio State University, Wexner
Medical Center, Columbus, Ohio
| | - Madeline Lewis
- Department of Surgery, The Ohio State University, Wexner
Medical Center, Columbus, Ohio
| | - Abhijoy Saha
- Department of Statistics, The Ohio State University,
Columbus, OH, USA
| | - Gayle Gordillo
- Department of Plastic Surgery, Indiana University School of
Medicine, Indianapolis, IN
- Department of Plastic Surgery, The Ohio State University,
Wexner Medical Center, Columbus, Ohio
| | - Savita Khanna
- Department of Surgery, The Ohio State University, Wexner
Medical Center, Columbus, Ohio
- Department of Plastic Surgery, Indiana University School of
Medicine, Indianapolis, IN
| |
Collapse
|
12
|
Wang Y, Jing Y, Ding L, Zhang X, Song Y, Chen S, Zhao X, Huang X, Pu Y, Wang Z, Ni Y, Hu Q. Epiregulin reprograms cancer-associated fibroblasts and facilitates oral squamous cell carcinoma invasion via JAK2-STAT3 pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:274. [PMID: 31234944 PMCID: PMC6591968 DOI: 10.1186/s13046-019-1277-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 06/12/2019] [Indexed: 01/12/2023]
Abstract
Background Local resident normal fibroblasts (NFs) are the major source of cancer-associated fibroblasts (CAFs), which are distinguishable from NFs by their tumor-supportive properties. However, the mechanism and the effects underlying the transition of NFs to CAFs in oral squamous cell carcinoma (OSCC) remain unclear. Methods Five pairs of matching primary NFs and CAFs derived from OSCC patients were sent for RNA sequencing. Epiregulin (EREG) expression was analyzed by IHC in fibroblasts from OSCC patients. The role of EREG in the NF-CAF transition and the consequential effects on OSCC progression were examined by upregulation/downregulation of EREG in NFs/CAFs both in vitro and in vivo. Results Here, we identified epiregulin (EREG) as the most remarkably upregulated gene in CAFs. High EREG expression in CAFs correlated with higher T stage, deeper invasion and inferior worst pattern of invasion (WPOI) in OSCC patients and predicted shorter overall survival. Overexpression of EREG in NFs activated the CAF phenotype. Mechanistically, the JAK2/STAT3 pathway was enhanced by EREG in parallel with increased IL-6 expression, which could be inhibited by the JAK2 inhibitor AG490. Recombinant IL-6 upregulated the JAK2/STAT3/EREG pathway in a feedback loop. Moreover, EREG-induced CAF activation promoted the epithelial-mesenchymal transition (EMT) necessary for migration and invasion, which was dependent on JAK2/STAT3 signaling and IL-6. In vivo, EREG expression in stroma fibroblasts promoted tumor growth with high stromal α-SMA, phospho-JAK2/STAT3, and IL-6 expression and upregulated EMT in HSC3 cells. Conclusions EREG is essential for the NF-CAF transformation needed to induce EMT of tumor cells in a JAK2-STAT3- and IL-6-dependent manner in OSCC. Electronic supplementary material The online version of this article (10.1186/s13046-019-1277-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yujia Wang
- Department of Oral & Maxillofacial Surgery Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China.,Central Laboratory Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China.,Department of Oral & Maxillofacial Surgery Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yue Jing
- Central Laboratory Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Liang Ding
- Central Laboratory Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Xiaoxin Zhang
- Central Laboratory Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Yuxian Song
- Central Laboratory Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Sheng Chen
- Department of Oral Pathology Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xingxing Zhao
- Department of Oral & Maxillofacial Surgery Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China.,Central Laboratory Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Xiaofeng Huang
- Department of Oral Pathology Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yumei Pu
- Department of Oral & Maxillofacial Surgery Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Zhiyong Wang
- Department of Oral & Maxillofacial Surgery Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Yanhong Ni
- Central Laboratory Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China.
| | - Qingang Hu
- Department of Oral & Maxillofacial Surgery Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China.
| |
Collapse
|
13
|
Gao X, Petricoin EF, Ward KR, Goldberg SR, Duane TM, Bonchev D, Arodz T, Diegelmann RF. Network proteomics of human dermal wound healing. Physiol Meas 2018; 39:124002. [PMID: 30524050 DOI: 10.1088/1361-6579/aaee19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The healing of wounds is critical in protecting the human body against environmental factors. The mechanisms involving protein expression during this complex physiological process have not been fully elucidated. APPROACH Here, we use reverse-phase protein microarrays (RPPA) involving 94 phosphoproteins to study tissue samples from tubes implanted in healing dermal wounds in seven human subjects tracked over two weeks. We compare the proteomic profiles to proteomes of controls obtained from skin biopsies from the same subjects. MAIN RESULTS Compared to previous proteomic studies of wound healing, our approach focuses on wound tissue instead of wound fluid, and has the sensitivity to go beyond measuring only highly abundant proteins. To study the temporal dynamics of networks involved in wound healing, we applied two network analysis methods that integrate the experimental results with prior knowledge about protein-protein physical and regulatory interactions, as well as higher-level biological processes and associated pathways. SIGNIFICANCE We uncovered densely connected networks of proteins that are up- or down-regulated during human wound healing, as well as their relationships to microRNAs and to proteins outside of our set of targets that we measured with proteomic microarrays.
Collapse
Affiliation(s)
- Xi Gao
- Department of Computer Science, School of Engineering, Virginia Commonwealth University, Richmond, VA, United States of America
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Richardson RJ. Parallels between vertebrate cardiac and cutaneous wound healing and regeneration. NPJ Regen Med 2018; 3:21. [PMID: 30416753 PMCID: PMC6220283 DOI: 10.1038/s41536-018-0059-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/12/2018] [Indexed: 12/11/2022] Open
Abstract
The cellular events that contribute to tissue healing of non-sterile wounds to the skin and ischaemic injury to internal organs such as the heart share remarkable similarities despite the differences between these injury types and organs. In adult vertebrates, both injuries are characterised by a complex series of overlapping events involving multiple different cell types and cellular interactions. In adult mammals both tissue-healing processes ultimately lead to the permanent formation of a fibrotic, collagenous scar, which can have varying effects on tissue function depending on the site and magnitude of damage. Extensive scarring in the heart as a result of a severe myocardial infarction contributes to ventricular dysfunction and the progression of heart failure. Some vertebrates such as adult zebrafish, however, retain a more embryonic capacity for scar-free tissue regeneration in many tissues including the skin and heart. In this review, the similarities and differences between these different types of wound healing are discussed, with special attention on recent advances in regenerative, non-scarring vertebrate models such as the zebrafish.
Collapse
Affiliation(s)
- Rebecca J Richardson
- School of Physiology, Pharmacology and Neuroscience, Faculty of Biomedical Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
15
|
Yang L, Zhang Z, Zhuo Y, Cui L, Li C, Li D, Zhang S, Cui N, Wang X, Gao H. Resveratrol alleviates sepsis-induced acute lung injury by suppressing inflammation and apoptosis of alveolar macrophage cells. Am J Transl Res 2018; 10:1961-1975. [PMID: 30093935 PMCID: PMC6079135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 10/10/2017] [Indexed: 06/08/2023]
Abstract
Sepsis is a major cause of death in intensive care units. The purpose of this study was to investigate the effect of resveratrol (RSV) on sepsis-induced acute lung injury (ALI). The underlying molecular mechanisms were deciphered by both in vitro and in vivo experiments. Polymicrobial sepsis was induced in C57BL/6 mice by cecal ligation and puncture (CLP). RSV pretreatment significantly attenuated CLP-induced acute lung injury, which was associated with enhanced expression of VEGF-B. The protective properties of RSV were assayed in lipopolysaccharide (LPS)-stimulated MH-S cells. We determine that RSV administration inhibited the increased production of TNF-α, IL-6, and IL-1β in LPS-stimulated MH-S cells, which was associated with inhibition of the nuclear factor-κB, P38, and ERK signaling pathways. We also provide evidence that RSV administration reduced LPS-induced apoptosis of MH-S cells by altering the unbalance of Bax/Bcl-2 and inhibiting LPS-induced autophagy. The inhibitory effects of RSV on cytokine levels and apoptosis of alveolar macrophages were both blocked by VEGF-B siRNA. Furthermore, RSV administration regulated LPS-induced C5aR and C5L2 expression, revealing an additional mechanism underlying RSV's anti-inflammatory and anti-apoptosis effects. Collectively, these results demonstrated that RSV was able to protect against sepsis-induced acute lung injury by activating the VEGF-B signaling pathway.
Collapse
Affiliation(s)
- Lei Yang
- Tianjin Institute of Acute Abdominal Disease of Integrated Traditional Chinese and Western MedicineTianjin 300100, China
| | - Zhen Zhang
- Graduate School of Tianjin Medical UniversityTianjin 300070, China
| | - Yuzhen Zhuo
- Tianjin Institute of Acute Abdominal Disease of Integrated Traditional Chinese and Western MedicineTianjin 300100, China
| | - Lihua Cui
- Tianjin Institute of Acute Abdominal Disease of Integrated Traditional Chinese and Western MedicineTianjin 300100, China
| | - Caixia Li
- Tianjin Institute of Acute Abdominal Disease of Integrated Traditional Chinese and Western MedicineTianjin 300100, China
| | - Dihua Li
- Tianjin Institute of Acute Abdominal Disease of Integrated Traditional Chinese and Western MedicineTianjin 300100, China
| | - Shukun Zhang
- Tianjin Institute of Acute Abdominal Disease of Integrated Traditional Chinese and Western MedicineTianjin 300100, China
| | - Naiqiang Cui
- Department of Surgery, Tianjin Hospital of Integrated Traditional Chinese and Western MedicineTianjin 300100, China
| | - Ximo Wang
- Graduate School of Tianjin Medical UniversityTianjin 300070, China
- Department of Surgery, Tianjin Hospital of Integrated Traditional Chinese and Western MedicineTianjin 300100, China
| | - Hongwei Gao
- Department of Anesthesiology, Perioperative & Pain Medicine, Brigham and Women’s Hospital, Harvard Medical SchoolBoston 02115, MA
| |
Collapse
|
16
|
Li J, Ghatak S, El Masry MS, Das A, Liu Y, Roy S, Lee RJ, Sen CK. Topical Lyophilized Targeted Lipid Nanoparticles in the Restoration of Skin Barrier Function following Burn Wound. Mol Ther 2018; 26:2178-2188. [PMID: 29802017 DOI: 10.1016/j.ymthe.2018.04.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/19/2018] [Accepted: 04/21/2018] [Indexed: 12/25/2022] Open
Abstract
Lyophilized keratinocyte-targeted nanocarriers (TLNκ) loaded with locked nucleic acid (LNA) modified anti-miR were developed for topical application to full thickness burn injury. TLNκ were designed to selectively deliver LNA-anti-miR-107 to keratinocytes using the peptide sequence ASKAIQVFLLAG. TLNκ employed DOTAP/DODAP combination pH-responsive lipid components to improve endosomal escape. To minimize interference of clearance by non-targeted cells, especially immune cells in the acute wound microenvironment, surface charge was neutralized. Lyophilization was performed to extend the shelf life of the lipid nanoparticles (LNPs). Encapsulation efficiency of anti-miR in lyophilized TLNκ was estimated to be 96.54%. Cargo stability of lyophilized TLNκ was tested. After 9 days of loading with anti-miR-210, TLNκ was effective in lowering abundance of the hypoxamiR miR-210 in keratinocytes challenged with hypoxia. Keratinocyte uptake of DiD-labeled TLNκ was selective and exceeded 90% within 4 hr. Topical application of hydrogel-dispersed lyophilized TLNκ encapsulating LNA anti-miR-107 twice a week significantly accelerated wound closure and restoration of skin barrier function. TLNκ/anti-miR-107 application depleted miR-107 and upregulated dicer expression, which accelerated differentiation of keratinocytes. Expression of junctional proteins such as claudin-1, loricrin, filaggrin, ZO-1, and ZO-2 were significantly upregulated following TLNκ/anti-miR-107 treatment. These LNPs are promising as topical therapeutic agents in the management of burn injury.
Collapse
Affiliation(s)
- Jilong Li
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Subhadip Ghatak
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA
| | - Mohamed S El Masry
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Department of General Surgery (Plastic Surgery Unit), Zagazig University, 44519, Egypt
| | - Amitava Das
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA
| | - Yang Liu
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Sashwati Roy
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA
| | - Robert J Lee
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA
| | - Chandan K Sen
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
17
|
Ceballos-Francisco D, Guardiola FA, Cordero H, Cuesta A, Esteban MÁ. Humoral immune parameters in serum of gilthead seabream (Sparus aurata L.) after induced skin injury. FISH & SHELLFISH IMMUNOLOGY 2018; 75:291-294. [PMID: 29448028 DOI: 10.1016/j.fsi.2018.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 02/01/2018] [Accepted: 02/07/2018] [Indexed: 06/08/2023]
Abstract
Skin lesions contribute to significant rates of morbidity and mortality in fish because they permit the entry of pathogens. Different seric immune parameters (IgM level, protease, antiprotease, peroxidase and lysozyme) and bactericidal activity (against Vibrio harveyi and Photobacterium damsealae) of gilthead seabream (Sparus aurata L.) specimens were evaluated after experimentally wounding fish in two body locations (above or below the lateral line). The results demonstrate that the level of several immune parameters present in fish serum (IgM, proteases, peroxidase and bactericidal activity) showed statistically significant variations depending on the site of the skin wound and the time post-wounding (from 0 to 7 days). However, other parameters (such as antiproteases or lysozyme) remained unaltered during the experiment and did not differs from the values recorded on control fish (non-wounded). The highest activities recorded coincided with the inflammatory healing phase. Moreover, many more significant variations were observed in fish wounded below the lateral line than in those wounded above the lateral line. The present results demonstrate the importance of skin integrity in the maintenance of fish body homeostasis.
Collapse
Affiliation(s)
- Diana Ceballos-Francisco
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100, Murcia, Spain
| | - Francisco A Guardiola
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100, Murcia, Spain; Fish Nutrition & Immunobiology Group, Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR), University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208, Porto, Portugal
| | - Héctor Cordero
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100, Murcia, Spain
| | - Alberto Cuesta
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100, Murcia, Spain
| | - M Ángeles Esteban
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100, Murcia, Spain.
| |
Collapse
|
18
|
Reprogramming of pro-inflammatory human macrophages to an anti-inflammatory phenotype by bile acids. Sci Rep 2018; 8:255. [PMID: 29321478 PMCID: PMC5762890 DOI: 10.1038/s41598-017-18305-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 12/04/2017] [Indexed: 12/11/2022] Open
Abstract
Cholestasis is caused by autoimmune reactions, drug-induced hepatotoxicity, viral infections of the liver and the obstruction of bile ducts by tumours or gallstones. Cholestatic conditions are associated with impaired innate and adaptive immunity, including alterations of the cellular functions of monocytes, macrophages, NK cells and T-cells. Bile acids act as signalling molecules, affecting lipopolysaccharide (LPS)-induced cytokine expression in primary human macrophages. The present manuscript investigates the impact of bile acids, such as taurolithocholic acid (TLC), on the transcriptome of human macrophages in the presence or absence of LPS. While TLC itself has almost no effect on gene expression under control conditions, this compound modulates the expression of 202 out of 865 transcripts in the presence of LPS. Interestingly, pathway analysis revealed that TLC specifically supressed the expression of genes involved in mediating pro-inflammatory effects, phagocytosis, interactions with pathogens and autophagy as well as the recruitment of immune cells, such as NK cells, neutrophils and T cells. These data indicate a broad influence of bile acids on inflammatory responses and immune functions in macrophages. These findings may contribute to the clinical observation that patients with cholestasis present a lack of response to bacterial or viral infections.
Collapse
|
19
|
Kunkemoeller B, Kyriakides TR. Redox Signaling in Diabetic Wound Healing Regulates Extracellular Matrix Deposition. Antioxid Redox Signal 2017; 27:823-838. [PMID: 28699352 PMCID: PMC5647483 DOI: 10.1089/ars.2017.7263] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SIGNIFICANCE Impaired wound healing is a major complication of diabetes, and can lead to development of chronic foot ulcers in a significant number of patients. Despite the danger posed by poor healing, very few specific therapies exist, leaving patients at risk of hospitalization, amputation, and further decline in overall health. Recent Advances: Redox signaling is a key regulator of wound healing, especially through its influence on the extracellular matrix (ECM). Normal redox signaling is disrupted in diabetes leading to several pathological mechanisms that alter the balance between reactive oxygen species (ROS) generation and scavenging. Importantly, pathological oxidative stress can alter ECM structure and function. CRITICAL ISSUES There is limited understanding of the specific role of altered redox signaling in the diabetic wound, although there is evidence that ROS are involved in the underlying pathology. FUTURE DIRECTIONS Preclinical studies of antioxidant-based therapies for diabetic wound healing have yielded promising results. Redox-based therapeutics constitute a novel approach for the treatment of wounds in diabetes patients that deserve further investigation. Antioxid. Redox Signal. 27, 823-838.
Collapse
Affiliation(s)
- Britta Kunkemoeller
- 1 Department of Pathology, Yale University School of Medicine , New Haven, Connecticut
- 2 Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine , New Haven, Connecticut
| | - Themis R Kyriakides
- 1 Department of Pathology, Yale University School of Medicine , New Haven, Connecticut
- 2 Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine , New Haven, Connecticut
- 3 Department of Biomedical Engineering, Yale University , New Haven, Connecticut
| |
Collapse
|
20
|
Gallego-Perez D, Pal D, Ghatak S, Malkoc V, Higuita-Castro N, Gnyawali S, Chang L, Liao WC, Shi J, Sinha M, Singh K, Steen E, Sunyecz A, Stewart R, Moore J, Ziebro T, Northcutt RG, Homsy M, Bertani P, Lu W, Roy S, Khanna S, Rink C, Sundaresan VB, Otero JJ, Lee LJ, Sen CK. Topical tissue nano-transfection mediates non-viral stroma reprogramming and rescue. NATURE NANOTECHNOLOGY 2017; 12:974-979. [PMID: 28785092 PMCID: PMC5814120 DOI: 10.1038/nnano.2017.134] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 06/09/2017] [Indexed: 05/08/2023]
Abstract
Although cellular therapies represent a promising strategy for a number of conditions, current approaches face major translational hurdles, including limited cell sources and the need for cumbersome pre-processing steps (for example, isolation, induced pluripotency). In vivo cell reprogramming has the potential to enable more-effective cell-based therapies by using readily available cell sources (for example, fibroblasts) and circumventing the need for ex vivo pre-processing. Existing reprogramming methodologies, however, are fraught with caveats, including a heavy reliance on viral transfection. Moreover, capsid size constraints and/or the stochastic nature of status quo approaches (viral and non-viral) pose additional limitations, thus highlighting the need for safer and more deterministic in vivo reprogramming methods. Here, we report a novel yet simple-to-implement non-viral approach to topically reprogram tissues through a nanochannelled device validated with well-established and newly developed reprogramming models of induced neurons and endothelium, respectively. We demonstrate the simplicity and utility of this approach by rescuing necrotizing tissues and whole limbs using two murine models of injury-induced ischaemia.
Collapse
Affiliation(s)
- Daniel Gallego-Perez
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
| | - Durba Pal
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
| | - Subhadip Ghatak
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
| | - Veysi Malkoc
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Natalia Higuita-Castro
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
| | - Surya Gnyawali
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
| | - Lingqian Chang
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, Ohio 43210, USA
| | - Wei-Ching Liao
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, Ohio 43210, USA
| | - Junfeng Shi
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Mithun Sinha
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
| | - Kanhaiya Singh
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
| | - Erin Steen
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
| | - Alec Sunyecz
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Richard Stewart
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
| | - Jordan Moore
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
| | - Thomas Ziebro
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Robert G. Northcutt
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Michael Homsy
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Paul Bertani
- Department of Electrical and Computer Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Wu Lu
- Department of Electrical and Computer Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Sashwati Roy
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
| | - Savita Khanna
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
| | - Cameron Rink
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
| | - Vishnu Baba Sundaresan
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Jose J. Otero
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Pathology, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210, USA
| | - L. James Lee
- Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, USA
- ;
| | - Chandan K. Sen
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, USA
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, Ohio 43210, USA
- ;
| |
Collapse
|
21
|
Das A, Datta S, Rhea B, Sinha M, Veeraragavan M, Gordillo G, Roy S. The Human Skeletal Muscle Transcriptome in Response to Oral Shilajit Supplementation. J Med Food 2017; 19:701-9. [PMID: 27414521 DOI: 10.1089/jmf.2016.0010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
UNLABELLED The objective of the present study ( clinicaltrials.gov NCT02026414) was to observe the effects of oral supplementation of a purified and standardized Shilajit extract on skeletal muscle adaptation in adult overweight/class I obese human subjects from the U.S. POPULATION Shilajit is a mineral pitch that oozes out of Himalayan rocks. The study design consisted of a baseline visit, followed by 8 weeks of 250 mg of oral Shilajit supplementation b.i.d., and additional 4 weeks of supplementation with exercise. At each visit, blood samples and muscle biopsies were collected for further analysis. Supplementation was well tolerated without any changes in blood glucose levels and lipid profile after 8 weeks of oral supplementation and the additional 4 weeks of oral supplementation with exercise. In addition, no changes were noted in creatine kinase and serum myoglobin levels after 8 weeks of oral supplementation and the additional 4 weeks of supplementation with exercise. Microarray analysis identified a cluster of 17 extracellular matrix (ECM)-related probe sets that were significantly upregulated in muscles following 8 weeks of oral supplementation compared with the expression at the baseline visit. This cluster included tenascin XB, decorin, myoferlin, collagen, elastin, fibrillin 1, and fibronectin 1. The differential expression of these genes was confirmed using quantitative real-time polymerase chain reaction (RT-PCR). The study provided maiden evidence that oral Shilajit supplementation in adult overweight/class I obese human subjects promoted skeletal muscle adaptation through upregulation of ECM-related genes that control muscle mechanotransduction properties, elasticity, repair, and regeneration.
Collapse
Affiliation(s)
- Amitava Das
- 1 Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine and Cell-Based Therapies and Comprehensive Wound Center, The Ohio State University , Wexner Medical Center, Columbus, Ohio, USA
| | - Soma Datta
- 1 Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine and Cell-Based Therapies and Comprehensive Wound Center, The Ohio State University , Wexner Medical Center, Columbus, Ohio, USA
| | - Brian Rhea
- 1 Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine and Cell-Based Therapies and Comprehensive Wound Center, The Ohio State University , Wexner Medical Center, Columbus, Ohio, USA
| | - Mithun Sinha
- 1 Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine and Cell-Based Therapies and Comprehensive Wound Center, The Ohio State University , Wexner Medical Center, Columbus, Ohio, USA
| | | | - Gayle Gordillo
- 3 Department of Plastic Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine and Cell-Based Therapies and Comprehensive Wound Center, The Ohio State University , Wexner Medical Center, Columbus, Ohio, USA
| | - Sashwati Roy
- 1 Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine and Cell-Based Therapies and Comprehensive Wound Center, The Ohio State University , Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
22
|
Das A, Ghatak S, Sinha M, Chaffee S, Ahmed NS, Parinandi NL, Wohleb ES, Sheridan JF, Sen CK, Roy S. Correction of MFG-E8 Resolves Inflammation and Promotes Cutaneous Wound Healing in Diabetes. THE JOURNAL OF IMMUNOLOGY 2016; 196:5089-100. [PMID: 27194784 DOI: 10.4049/jimmunol.1502270] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 04/18/2016] [Indexed: 12/15/2022]
Abstract
Milk fat globule epidermal growth factor-factor 8 (MFG-E8) is a peripheral glycoprotein that acts as a bridging molecule between the macrophage and apoptotic cells, thus executing a pivotal role in the scavenging of apoptotic cells from affected tissue. We have previously reported that apoptotic cell clearance activity or efferocytosis is compromised in diabetic wound macrophages. In this work, we test the hypothesis that MFG-E8 helps resolve inflammation, supports angiogenesis, and accelerates wound closure. MFG-E8(-/-) mice displayed impaired efferocytosis associated with exaggerated inflammatory response, poor angiogenesis, and wound closure. Wound macrophage-derived MFG-E8 was recognized as a critical driver of wound angiogenesis. Transplantation of MFG-E8(-/-) bone marrow to MFG-E8(+/+) mice resulted in impaired wound closure and compromised wound vascularization. In contrast, MFG-E8(-/-) mice that received wild-type bone marrow showed improved wound closure and improved wound vascularization. Hyperglycemia and exposure to advanced glycated end products inactivated MFG-E8, recognizing a key mechanism that complicates diabetic wound healing. Diabetic db/db mice suffered from impaired efferocytosis accompanied with persistent inflammation and slow wound closure. Topical recombinant MFG-E8 induced resolution of wound inflammation, improvements in angiogenesis, and acceleration of closure, upholding the potential of MFG-E8-directed therapeutics in diabetic wound care.
Collapse
Affiliation(s)
- Amitava Das
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Subhadip Ghatak
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Mithun Sinha
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Scott Chaffee
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Noha S Ahmed
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Narasimham L Parinandi
- Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, OH 43210; and
| | - Eric S Wohleb
- Division of Biosciences, The Ohio State University, Columbus, OH 43210
| | - John F Sheridan
- Division of Biosciences, The Ohio State University, Columbus, OH 43210
| | - Chandan K Sen
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Sashwati Roy
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210;
| |
Collapse
|
23
|
Anderson AE, Galko MJ. Will the wound-healing field earn its wings? Exp Dermatol 2016; 23:809-10. [PMID: 25040854 DOI: 10.1111/exd.12498] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2014] [Indexed: 12/25/2022]
Abstract
In a recently published issue of Experimental Dermatology, Dr. Nuria Paricio and colleagues review recent advances using the fruit fly, Drosophila melanogaster, as a wound-healing model. They describe many of the advantages of the fly model for gene discovery and functional analysis, highlighting its particular strengths and limitations for studies of wound healing. This commentary assumes that dermatologist-scientists and fly wound-healing researchers share a common field-wide goal of discovering all of the clinically relevant wound-healing genes and understanding in molecular detail how those genes work. We ask: how can we cooperate to achieve this shared goal?
Collapse
Affiliation(s)
- Aimee E Anderson
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
24
|
Powers CJ, Dickerson R, Zhang SW, Rink C, Roy S, Sen CK. Human cerebrospinal fluid microRNA: temporal changes following subarachnoid hemorrhage. Physiol Genomics 2016; 48:361-6. [PMID: 26945012 DOI: 10.1152/physiolgenomics.00052.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 03/01/2016] [Indexed: 02/01/2023] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is a devastating form of hemorrhagic stroke with 30-day mortality between 33 and 45%. Delayed cerebral ischemia (DCI) is the chief cause of morbidity and mortality in patients who survive the initial aSAH. DCI accounts for almost 50% of deaths in patients surviving to treatment of the ruptured aneurysm. The mechanisms for brain injury after aSAH and the brain's response to this injury are not fully understood in humans. MicroRNAs (miRs) are 22- to 25-nucleotide single-stranded RNA molecules that inhibit the expression of specific messenger RNA targets. In this work, miR profiling of human cerebrospinal fluid from eight patients after aSAH was performed daily for 10 days with the goal of identifying changes in miR abundance. Using the nanoString nCounter Expression Assay, we identified two specific clusters of miR that were differentially regulated over time. Quantitative RT-PCR was performed on select miRs from each cluster. The first cluster contained miRs known to be present in blood and decreased in abundance over time. miRs in this group include miR-92a and let-7b. The second cluster contained several poorly characterized miRs that increased in abundance over time. miRs in this group included miR-491. This second cluster of miRs may be released into the CSF by the brain itself as a result of the initial SAH. Temporal changes in the abundance of specific miRs in human CSF after aSAH may provide novel insight into the role of miRs in brain injury and the brain's response.
Collapse
Affiliation(s)
- Ciarán J Powers
- Department of Neurological Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio; and
| | - Ryan Dickerson
- Department of Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio
| | - Stacey W Zhang
- Department of Neurological Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio; and
| | - Cameron Rink
- Department of Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio
| | - Sashwati Roy
- Department of Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio
| | - Chandan K Sen
- Department of Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
25
|
Abstract
Diabetic neuropathies (DNs) are one of the most prevalent chronic complications of diabetes and a major cause of disability, high mortality, and poor quality of life. Given the complex anatomy of the peripheral nervous system and types of fiber dysfunction, DNs have a wide spectrum of clinical manifestations. The treatment of DNs continues to be challenging, likely due to the complex pathogenesis that involves an array of systemic and cellular imbalances in glucose and lipids metabolism. These lead to the activation of various biochemical pathways, including increased oxidative/nitrosative stress, activation of the polyol and protein kinase C pathways, activation of polyADP ribosylation, and activation of genes involved in neuronal damage, cyclooxygenase-2 activation, endothelial dysfunction, altered Na(+)/K(+)-ATPase pump function, impaired C-peptide-related signaling pathways, endoplasmic reticulum stress, and low-grade inflammation. This review summarizes current evidence regarding the role of low-grade inflammation as a potential therapeutic target for DNs.
Collapse
Affiliation(s)
- Rodica Pop-Busui
- Department of Internal Medicine, Division of Metabolism, Metabolism Endocrinology and Diabetes, University of Michigan, 5329 Brehm Tower, 1000 Wall Street, Ann Arbor, MI, 48105, USA.
| | - Lynn Ang
- Department of Internal Medicine, Division of Metabolism, Metabolism Endocrinology and Diabetes, University of Michigan, 5329 Brehm Tower, 1000 Wall Street, Ann Arbor, MI, 48105, USA.
| | - Crystal Holmes
- The Division of Metabolism, Endocrinology and Diabetes, Dominos Farms, Lobby C, Suite 1300 24 Frank Lloyd Wright Drive, PO Box 451, Ann Arbor, MI, 48106-0451, USA.
| | - Katherine Gallagher
- Department of Surgery, Section of Vascular Surgery, University of Michigan Health System, 1500 East Medical Center Dr, SPC 5867, Ann Arbor, MI, 48109, USA.
| | - Eva L Feldman
- Department of Neurology, University of Michigan, 5017 AATBSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
26
|
Sen CK, Ghatak S. miRNA control of tissue repair and regeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2629-40. [PMID: 26056933 DOI: 10.1016/j.ajpath.2015.04.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 03/30/2015] [Accepted: 04/02/2015] [Indexed: 12/21/2022]
Abstract
Tissue repair and regeneration rely on the function of miRNA, molecular silencers that enact post-transcriptional gene silencing of coding genes. Disruption of miRNA homeostasis is developmentally lethal, indicating that fetal tissue development is tightly controlled by miRNAs. Multiple critical facets of adult tissue repair are subject to control by miRNAs, as well. Sources of cell pool for tissue repair and regeneration are diverse and provided by processes including cellular dedifferentiation, transdifferentiation, and reprogramming. Each of these processes is regulated by miRNAs. Furthermore, induced pluripotency may be achieved by miRNA-based strategies independent of transcription factor manipulation. The observation that miRNA does not integrate into the genome makes miRNA-based therapeutic strategies translationally valuable. Tools to manipulate cellular and tissue miRNA levels include mimics and inhibitors that may be specifically targeted to cells of interest at the injury site. Here, we discuss the extraordinary importance of miRNAs in tissue repair and regeneration based on emergent reports and rapid advances in miRNA-based therapeutics.
Collapse
Affiliation(s)
- Chandan K Sen
- Center for Regenerative Medicine and Cell-Based Therapies and the Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio.
| | - Subhadip Ghatak
- Center for Regenerative Medicine and Cell-Based Therapies and the Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
27
|
Genome-wide expression analysis of wounded skin reveals novel genes involved in angiogenesis. Angiogenesis 2015; 18:361-71. [DOI: 10.1007/s10456-015-9472-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 05/20/2015] [Indexed: 01/29/2023]
|
28
|
Barrier Function of the Repaired Skin Is Disrupted Following Arrest of Dicer in Keratinocytes. Mol Ther 2015; 23:1201-1210. [PMID: 25896246 DOI: 10.1038/mt.2015.65] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 04/07/2015] [Indexed: 02/07/2023] Open
Abstract
Tissue injury transiently silences miRNA-dependent posttranscriptional gene silencing in its effort to unleash adult tissue repair. Once the wound is closed, miRNA biogenesis is induced averting neoplasia. In this work, we report that Dicer plays an important role in reestablishing the barrier function of the skin post-wounding via a miRNA-dependent mechanism. MicroRNA expression profiling of skin and wound-edge tissue revealed global upregulation of miRNAs following wound closure at day 14 post-wounding with significant induction of Dicer expression. Barrier function of the skin, as measured by trans-epidermal water loss, was compromised in keratinocyte-specific conditional (K14/Lox-Cre) Dicer-ablated mice because of malformed cornified epithelium lacking loricrin expression. Studies on human keratinocytes recognized that loricrin expression was inversely related to the expression of the cyclin-dependent kinase inhibitor p21(Waf1/Cip1). Compared to healthy epidermis, wound-edge keratinocytes from Dicer-ablated skin epidermis revealed elevated p21(Waf1/Cip1) expression. Adenoviral and pharmacological suppression of p21(Waf1/Cip1) in keratinocyte-specific conditional Dicer-ablated mice improved wound healing indicating a role of Dicer in the suppression of p21(Waf1/Cip1). This work upholds p21(Waf1/Cip1) as a druggable target to restore barrier function of skin suffering from loss of Dicer function as would be expected in diabetes and other forms of oxidant insult.
Collapse
|
29
|
Peake MA, Caley M, Giles PJ, Wall I, Enoch S, Davies LC, Kipling D, Thomas DW, Stephens P. Identification of a transcriptional signature for the wound healing continuum. Wound Repair Regen 2015; 22:399-405. [PMID: 24844339 PMCID: PMC4230470 DOI: 10.1111/wrr.12170] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 02/19/2014] [Indexed: 11/28/2022]
Abstract
There is a spectrum/continuum of adult human wound healing outcomes ranging from the enhanced (nearly scarless) healing observed in oral mucosa to scarring within skin and the nonhealing of chronic skin wounds. Central to these outcomes is the role of the fibroblast. Global gene expression profiling utilizing microarrays is starting to give insight into the role of such cells during the healing process, but no studies to date have produced a gene signature for this wound healing continuum. Microarray analysis of adult oral mucosal fibroblast (OMF), normal skin fibroblast (NF), and chronic wound fibroblast (CWF) at 0 and 6 hours post-serum stimulation was performed. Genes whose expression increases following serum exposure in the order OMF < NF < CWF are candidates for a negative/impaired healing phenotype (the dysfunctional healing group), whereas genes with the converse pattern are potentially associated with a positive/preferential healing phenotype (the enhanced healing group). Sixty-six genes in the enhanced healing group and 38 genes in the dysfunctional healing group were identified. Overrepresentation analysis revealed pathways directly and indirectly associated with wound healing and aging and additional categories associated with differentiation, development, and morphogenesis. Knowledge of this wound healing continuum gene signature may in turn assist in the therapeutic assessment/treatment of a patient's wounds.
Collapse
Affiliation(s)
- Matthew A Peake
- Wound Biology Group, Cardiff Institute of Tissue Engineering and Repair, Tissue Engineering and Reparative Dentistry, School of Dentistry
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Tsai TC, Tung YT, Kuo YH, Liao JW, Tsai HC, Chong KY, Chen HL, Chen CM. Anti-inflammatory effects of Antrodia camphorata, a herbal medicine, in a mouse skin ischemia model. JOURNAL OF ETHNOPHARMACOLOGY 2015; 159:113-121. [PMID: 25449461 DOI: 10.1016/j.jep.2014.11.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/23/2014] [Accepted: 11/07/2014] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL EVIDENCE Antrodia camphorata, a highly valued polypore mushroom native only to Taiwan, has been traditionally used as a medicine for anti-inflammation. AIM OF THE STUDY In this study, anti-inflammatory effects of Antrodia camphorata (AC) and its active compound, ergostatrien-3β-ol (ST1), were investigated in a mouse skin ischemia model induced by skin flap surgery on the dorsal skin. MATERIALS AND METHODS A U-shaped flap was elevated on the dorsal skin of the nine-week-old male mice. Mice were randomly assigned to six groups for treatment (n=6) including normal skin/propylene glycol (PG), surgical skin flap/PG, solid-state-cultured AC (S/AC), wood-cultured AC (W/AC), high-dose ST1 (H-ST1), low-dose ST1 (L-ST1). Antrodia camphorata was dissolved in 25μL PG and smeared on the skin flap every six hours for 24h. At the end of the experiment, each mouse was anesthetized, and skin tissues were collected from their back for histopathological analysis, extracting RNA and protein according to our previous reports. RESULTS Skin-flap-induced ischemia damage significantly increased the expression of the iNOS, COX2, and IL-6 proteins and decreased the expression of IκB protein. In addition, focal, moderate coagulative necrosis with inflammatory cell infiltration was found in the epidermis, and moderate inflammatory cells and necrosis with slight edema was noted in the sub-dermis at 24h after skin flap surgery. However, treatment with solid-state-cultured or wood-cultured AC, or with its derived ST1 active compound, significantly reduced the necrosis and inflammatory cell infiltration in both the epidermis and sub-dermis of the skin flap. The treatments also reduced the inflammatory response by decreasing the expression of inflammation-related genes including iNOS, IL-6, TNF-α, and NF-κB, as shown by changes in RNA and protein expression, when compared with the surgical skin flap procedure alone. CONCLUSIONS These results demonstrated that methanolic extracts of wood-cultured fruiting bodies and solid-state-cultured mycelia from Antrodia camphorata have excellent anti-inflammatory activities and thus have great potential as an addition for hydrocolloid dressings.
Collapse
Affiliation(s)
- Tung-Chou Tsai
- Department of Life Sciences, and Agricultural Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
| | - Yu-Tang Tung
- Department of Life Sciences, and Agricultural Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
| | - Yueh-Hsiung Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 404, Taiwan; Department of Biotechnology, Asia University, Taichung 413, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Taichung 402, Taiwan
| | - Hsin-Chung Tsai
- Department of Sugery, Taichung Hospital, Ministry of Health and Welfare, Taichung 403, Taiwan
| | - Kowit-Yu Chong
- Department of Medical Biotechnology and Laboratory Science, Molecular Medicine Research Center, Chang Gung University, Tao-Yuan 333, Taiwan
| | - Hsiao-Ling Chen
- Department of Bioresources, Da-Yeh University, Changhua 515, Taiwan.
| | - Chuan-Mu Chen
- Department of Life Sciences, and Agricultural Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan; Rong-Hsing Translational Medicine Center, and the iEGG center, National Chung Hsing University, Taichung 402, Taiwan.
| |
Collapse
|
31
|
Abstract
microRNAs (miRNAs) are small noncoding RNA molecules which play pivotal roles in wound healing. The increased expression of certain genes and expression of some others represent a key component of the wound biology and are largely under the regulation of naturally occurring miRNAs. Understanding the dysregulated miRNAs in chronic wound biology will therefore enable the development of newer therapies. This chapter focuses on the miRNAs that can be potentially targeted for improving skin wound healing and the challenges in miRNA therapy, including considerations in miRNA target identification and delivery.
Collapse
|
32
|
Elgharably H, Ganesh K, Dickerson J, Khanna S, Abas M, Ghatak PD, Dixit S, Bergdall V, Roy S, Sen CK. A modified collagen gel dressing promotes angiogenesis in a preclinical swine model of chronic ischemic wounds. Wound Repair Regen 2014; 22:720-9. [PMID: 25224310 PMCID: PMC4380279 DOI: 10.1111/wrr.12229] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/04/2014] [Indexed: 01/23/2023]
Abstract
We recently performed proteomic characterization of a modified collagen gel (MCG) dressing and reported promising effects of the gel in healing full-thickness excisional wounds. In this work, we test the translational relevance of our aforesaid findings by testing the dressing in a swine model of chronic ischemic wounds recently reported by our laboratory. Full-thickness excisional wounds were established in the center of bipedicle ischemic skin flaps on the backs of animals. Ischemia was verified by laser Doppler imaging, and MCG was applied to the test group of wounds. Seven days post wounding, macrophage recruitment to the wound was significantly higher in MCG-treated ischemic wounds. In vitro, MCG up-regulated expression of Mrc-1 (a reparative M2 macrophage marker) and induced the expression of anti-inflammatory cytokine interleukin (IL)-10 and of fibroblast growth factor-basic (β-FGF). An increased expression of CCR2, an M2 macrophage marker, was noted in the macrophages from MCG treated wounds. Furthermore, analyses of wound tissues 7 days post wounding showed up-regulation of transforming growth factor-β, vascular endothelial growth factor, von Willebrand's factor, and collagen type I expression in MCG-treated ischemic wounds. At 21 days post wounding, MCG-treated ischemic wounds displayed higher abundance of proliferating endothelial cells that formed mature vascular structures and increased blood flow to the wound. Fibroblast count was markedly higher in MCG-treated ischemic wound-edge tissue. In addition, MCG-treated wound-edge tissues displayed higher abundance of mature collagen with increased collagen type I : III deposition. Taken together, MCG helped mount a more robust inflammatory response that resolved in a timely manner, followed by an enhanced proliferative phase, angiogenic outcome, and postwound tissue remodeling. Findings of the current study warrant clinical testing of MCG in a setting of ischemic chronic wounds.
Collapse
Affiliation(s)
- Haytham Elgharably
- Department of Surgery, Davis Heart & Lung Research Institute, Center for Regenerative Medicine and Cell Based Therapies, Comprehensive Wound Center, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Hong S, Tian H, Lu Y, Laborde JM, Muhale FA, Wang Q, Alapure BV, Serhan CN, Bazan NG. Neuroprotectin/protectin D1: endogenous biosynthesis and actions on diabetic macrophages in promoting wound healing and innervation impaired by diabetes. Am J Physiol Cell Physiol 2014; 307:C1058-67. [PMID: 25273880 DOI: 10.1152/ajpcell.00270.2014] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Dysfunction of macrophages (MΦs) in diabetic wounds impairs the healing. MΦs produce anti-inflammatory and pro-resolving neuroprotectin/protectin D1 (NPD1/PD1, 10R,17S-dihydroxy-docosa-4Z,7Z,11E,13E,15Z,19Z-hexaenoic acid); however, little is known about endogenous NPD1 biosynthesis by MΦs and the actions of NPD1 on diabetic MΦ functions in diabetic wound healing. We used an excisional skin wound model of diabetic mice, MΦ depletion, MΦs isolated from diabetic mice, and mass spectrometry-based targeted lipidomics to study the time course progression of NPD1 levels in wounds, the roles of MΦs in NPD1 biosynthesis, and NPD1 action on diabetic MΦ inflammatory activities. We also investigated the healing, innervation, chronic inflammation, and oxidative stress in diabetic wounds treated with NPD1 or NPD1-modulated MΦs from diabetic mice. Injury induced endogenous NPD1 biosynthesis in wounds, but diabetes impeded NPD1 formation. NPD1 was mainly produced by MΦs. NPD1 enhanced wound healing and innervation in diabetic mice and promoted MΦs functions that accelerated these processes. The underlying mechanisms for these actions of NPD1 or NPD1-modulated MΦs involved 1) attenuating MΦ inflammatory activities and chronic inflammation and oxidative stress after acute inflammation in diabetic wound, and 2) increasing MΦ production of IL10 and hepatocyte growth factor. Taken together, NPD1 appears to be a MΦs-produced factor that accelerates diabetic wound healing and promotes MΦ pro-healing functions in diabetic wounds. Decreased NPD1 production in diabetic wound is associated with impaired healing. This study identifies a new molecular target that might be useful in development of more effective therapeutics based on NPD1 and syngeneic diabetic MΦs for treatment of diabetic wounds.
Collapse
Affiliation(s)
- Song Hong
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana; Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, Louisiana;
| | - Haibin Tian
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Yan Lu
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - James Monroe Laborde
- Department of Orthopedic Surgery, Louisiana State University Health Sciences Center, New Orleans, Louisiana; and
| | - Filipe A Muhale
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Quansheng Wang
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Bhagwat V Alapure
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital; Department of Anaesthesia (Biochemistry and Molecular Pharmacology), Harvard Medical School; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana; Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| |
Collapse
|
34
|
Abstract
Epiregulin is a 46-amino acid protein that belongs to the epidermal growth factor (EGF) family of peptide hormones. Epiregulin binds to the EGF receptor (EGFR/ErbB1) and ErbB4 (HER4) and can stimulate signaling of ErbB2 (HER2/Neu) and ErbB3 (HER3) through ligand-induced heterodimerization with a cognate receptor. Epiregulin possesses a range of functions in both normal physiologic states as well as in pathologic conditions. Epiregulin contributes to inflammation, wound healing, tissue repair, and oocyte maturation by regulating angiogenesis and vascular remodeling and by stimulating cell proliferation. Deregulated epiregulin activity appears to contribute to the progression of a number of different malignancies, including cancers of the bladder, stomach, colon, breast, lung, head and neck, and liver. Therefore, epiregulin and the elements of the EGF/ErbB signaling network that lie downstream of epiregulin appear to be good targets for therapeutic intervention.
Collapse
|
35
|
Epiregulin: roles in normal physiology and cancer. Semin Cell Dev Biol 2014; 28:49-56. [PMID: 24631357 DOI: 10.1016/j.semcdb.2014.03.005] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 02/24/2014] [Accepted: 03/03/2014] [Indexed: 12/13/2022]
Abstract
Epiregulin is a 46-amino acid protein that belongs to the epidermal growth factor (EGF) family of peptide hormones. Epiregulin binds to the EGF receptor (EGFR/ErbB1) and ErbB4 (HER4) and can stimulate signaling of ErbB2 (HER2/Neu) and ErbB3 (HER3) through ligand-induced heterodimerization with a cognate receptor. Epiregulin possesses a range of functions in both normal physiologic states as well as in pathologic conditions. Epiregulin contributes to inflammation, wound healing, tissue repair, and oocyte maturation by regulating angiogenesis and vascular remodeling and by stimulating cell proliferation. Deregulated epiregulin activity appears to contribute to the progression of a number of different malignancies, including cancers of the bladder, stomach, colon, breast, lung, head and neck, and liver. Therefore, epiregulin and the elements of the EGF/ErbB signaling network that lie downstream of epiregulin appear to be good targets for therapeutic intervention.
Collapse
|
36
|
Das A, Ganesh K, Khanna S, Sen CK, Roy S. Engulfment of apoptotic cells by macrophages: a role of microRNA-21 in the resolution of wound inflammation. THE JOURNAL OF IMMUNOLOGY 2014; 192:1120-9. [PMID: 24391209 DOI: 10.4049/jimmunol.1300613] [Citation(s) in RCA: 237] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
At an injury site, efficient clearance of apoptotic cells by wound macrophages or efferocytosis is a prerequisite for the timely resolution of inflammation. Emerging evidence indicates that microRNA-21 (miR-21) may regulate the inflammatory response. In this work, we sought to elucidate the significance of miR-21 in the regulation of efferocytosis-mediated suppression of innate immune response, a key process implicated in resolving inflammation following injury. An increased expression of inducible miR-21 was noted in postefferocytotic peripheral blood monocyte-derived macrophages. Such induction of miR-21 was associated with silencing of its target genes PTEN and PDCD4. Successful efferocytosis of apoptotic cells by monocyte-derived macrophages resulted in the suppression of LPS-induced NF-κB activation and TNF-α expression. Interestingly, bolstering of miR-21 levels alone, using miR mimic, resulted in significant suppression of LPS-induced TNF-α expression and NF-κB activation. We report that efferocytosis-induced miR-21, by silencing PTEN and GSK3β, tempers the LPS-induced inflammatory response. Macrophage efferocytosis is known to trigger the release of anti-inflammatory cytokine IL-10. This study demonstrates that following successful efferocytosis, miR-21 induction in macrophages silences PDCD4, favoring c-Jun-AP-1 activity, which in turn results in elevated production of anti-inflammatory IL-10. In summary, this work provides direct evidence implicating miRNA in the process of turning on an anti-inflammatory phenotype in the postefferocytotic macrophage. Elevated macrophage miR-21 promotes efferocytosis and silences target genes PTEN and PDCD4, which in turn accounts for a net anti-inflammatory phenotype. Findings of this study highlight the significance of miRs in the resolution of wound inflammation.
Collapse
Affiliation(s)
- Amitava Das
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine and Cell Based Therapies and Comprehensive Wound Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | | | | | | | | |
Collapse
|
37
|
Keratin 16 regulates innate immunity in response to epidermal barrier breach. Proc Natl Acad Sci U S A 2013; 110:19537-42. [PMID: 24218583 DOI: 10.1073/pnas.1309576110] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mutations in the type I keratin 16 (Krt16) and its partner type II keratin 6 (Krt6a, Krt6b) cause pachyonychia congenita (PC), a disorder typified by dystrophic nails, painful hyperkeratotic calluses in glabrous skin, and lesions involving other epithelial appendages. The pathophysiology of these symptoms and its relationship to settings in which Krt16 and Krt6 are induced in response to epidermal barrier stress are poorly understood. We report that hyperkeratotic calluses arising in the glabrous skin of individuals with PC and Krt16 null mice share a gene expression signature enriched in genes involved in inflammation and innate immunity, in particular damage-associated molecular patterns. Transcriptional hyper-activation of damage-associated molecular pattern genes occurs following de novo chemical or mechanical irritation to ear skin and in spontaneously arising skin lesions in Krt16 null mice. Genome-wide expression analysis of normal mouse tail skin and benign proliferative lesions reveals a tight, context-dependent coregulation of Krt16 and Krt6 with genes involved in skin barrier maintenance and innate immunity. Our results uncover a role for Krt16 in regulating epithelial inflammation that is relevant to genodermatoses, psoriasis, and cancer and suggest a avenue for the therapeutic management of PC and related disorders.
Collapse
|
38
|
Arodz T, Bonchev D, Diegelmann RF. A Network Approach to Wound Healing. Adv Wound Care (New Rochelle) 2013; 2:499-509. [PMID: 24527361 DOI: 10.1089/wound.2012.0386] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 12/22/2012] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE The wound healing process is well-understood on the cellular and tissue level; however, its complex molecular mechanisms are not yet uncovered in their entirety. Viewing wounds as perturbed molecular networks provides the tools for analyzing and optimizing the healing process. It helps to answer specific questions that lead to better understanding of the complexity of the process. What are the molecular pathways involved in wound healing? How do these pathways interact with each other during the different stages of wound healing? Is it possible to grasp the entire mechanism of regulatory interactions in the healing of a wound? APPROACH Networks are structures composed of nodes connected by links. A network describing the state of a cell taking part in the healing process may contain nodes representing genes, proteins, microRNAs, metabolites, and drug molecules. The links connecting nodes represent interactions such as binding, regulation, co-expression, chemical reaction, and others. Both nodes and links can be weighted by numbers related to molecular concentration and the intensity of intermolecular interactions. Proceeding from data and from molecular profiling experiments, different types of networks are built to characterize the stages of the healing process. Network nodes having a higher degree of connectivity and centrality usually play more important roles for the functioning of the system they describe. RESULTS We describe here the algorithms and software packages for building, manipulating and analyzing networks proceeding from information available from a literature or database search or directly extracted from experimental gene expression, metabolic, and proteomic data. Network analysis identifies genes/proteins most differentiated during the healing process, and their organization in functional pathways or modules, and their distribution into gene ontology categories of biological processes, molecular functions, and cellular localization. We provide an example of how network analysis can be used to reach better understanding of regulation of key wound healing mediators and microRNAs that regulate them. INNOVATION Univariate statistical tests widely used in clinical studies are not enough to improve understanding and optimize the processes of wound healing. Network methods of analysis of patients "omics" data, such as transcriptoms, proteomes, and others can provide a better insight into the healing processes and help in development of better treatment practices. We review several articles that are examples of this emergent approach to the study of wound healing. CONCLUSION Network analysis has the potential to considerably contribute to the better understanding of the molecular mechanisms of wound healing and to the discovery of means to control and optimize that process.
Collapse
Affiliation(s)
- Tomasz Arodz
- Department of Computer Science, Virginia Commonwealth University, Richmond, Virginia
- VCU Reanimation, Engineering, and Science Center, Virginia Commonwealth University, Richmond, Virginia
| | - Danail Bonchev
- VCU Reanimation, Engineering, and Science Center, Virginia Commonwealth University, Richmond, Virginia
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia
| | - Robert F. Diegelmann
- VCU Reanimation, Engineering, and Science Center, Virginia Commonwealth University, Richmond, Virginia
- Department of Biochemistry & Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
39
|
Kashpur O, LaPointe D, Ambady S, Ryder EF, Dominko T. FGF2-induced effects on transcriptome associated with regeneration competence in adult human fibroblasts. BMC Genomics 2013; 14:656. [PMID: 24066673 PMCID: PMC3849719 DOI: 10.1186/1471-2164-14-656] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 09/24/2013] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Adult human fibroblasts grown in low oxygen and with FGF2 supplementation have the capacity to tip the healing outcome of skeletal muscle injury - by favoring regeneration response in vivo over scar formation. Here, we compare the transcriptomes of control adult human dermal fibroblasts and induced regeneration-competent (iRC) fibroblasts to identify transcriptional changes that may be related to their regeneration competence. RESULTS We identified a unique gene-expression profile that characterizes FGF2-induced iRC fibroblast phenotype. Significantly differentially expressed genes due to FGF2 treatment were identified and analyzed to determine overrepresented Gene Ontology terms. Genes belonging to extracellular matrix components, adhesion molecules, matrix remodelling, cytoskeleton, and cytokines were determined to be affected by FGF2 treatment. CONCLUSIONS Transcriptome analysis comparing control adult human fibroblasts with FGF2-treated fibroblasts identified functional groups of genes that reflect transcriptional changes potentially contributing to their regeneration competence. This comparative transcriptome analysis should contribute new insights into genes that characterize cells with greater regenerative potential.
Collapse
Affiliation(s)
- Olga Kashpur
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA 01609, USA.
| | | | | | | | | |
Collapse
|
40
|
Yang L, Engeland CG, Cheng B. Social isolation impairs oral palatal wound healing in sprague-dawley rats: a role for miR-29 and miR-203 via VEGF suppression. PLoS One 2013; 8:e72359. [PMID: 23951316 PMCID: PMC3739786 DOI: 10.1371/journal.pone.0072359] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 07/15/2013] [Indexed: 12/23/2022] Open
Abstract
Objective To investigate the effects of social isolation on oral mucosal healing in rats, and to determine if wound-associated genes and microRNAs (miRNAs) may contribute to this response. Methods Rats were group housed or socially isolated for 4 weeks before a 3.5 mm wound was placed on the hard oral palate. Wound closure was assessed daily and tissues were collected for determination of gene expression levels and miRNAs (i.e., miR-29a,b,c and miR-203). The predicted target of these microRNAs (i.e., vascular endothelial growth factor A, VEGFA) was functionally validated. Results Social isolation stress delayed the healing process of oral palatal mucosal wounds in rats. Lower mRNA levels of interleukin-1β (IL1β), macrophage inflammatory protein-1α (MIP1α), fibroblast growth factor 7 (FGF7), and VEGFA were found in the biopsied tissues of isolated animals on days 1 and/or 3 post-wounding. Intriguingly, the isolated rats persistently exhibited higher levels of miR-29 family members and miR-203. Our results confirmed that VEGFA is a direct target of these miRNAs, as both miR-29a,c and miR-203 strongly and specifically suppressed endogenous VEGFA expression in vitro. Conclusions This study in rats demonstrates for the first time that social isolation delays oral mucosal healing, and suggests a potential role for healing-associated gene and miRNA interactions during this process via modulation of VEGF expression.
Collapse
Affiliation(s)
- Linglan Yang
- Department of Oral Medicine, the Affiliated Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| | - Christopher G. Engeland
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Women, Child, Family Health Science, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Bin Cheng
- Department of Oral Medicine, the Affiliated Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
- * E-mail:
| |
Collapse
|
41
|
Haemmerle M, Keller T, Egger G, Schachner H, Steiner CW, Stokic D, Neumayer C, Brown MK, Kerjaschki D, Hantusch B. Enhanced lymph vessel density, remodeling, and inflammation are reflected by gene expression signatures in dermal lymphatic endothelial cells in type 2 diabetes. Diabetes 2013; 62:2509-29. [PMID: 23423575 PMCID: PMC3712036 DOI: 10.2337/db12-0844] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Type 2 diabetes is associated with microvascular damage that causes frequent infections in the skin and chronic ulcers as a result of impaired wound healing. To trace the pathological changes, we performed a comprehensive analysis of lymphatic vessels in the skin of type 2 diabetic versus nondiabetic patients. The dermis revealed enhanced lymphatic vessel density, and transcriptional profiling of ex vivo isolated lymphatic endothelial cells (LECs) identified 160 genes differentially expressed between type 2 diabetic and nondiabetic LECs. Bioinformatic analysis of deregulated genes uncovered sets functionally related to inflammation, lymphatic vessel remodeling, lymphangiogenesis, and lipid and small molecule transport. Furthermore, we traced CD68(+) macrophage accumulation and concomitant upregulation of tumor necrosis factor-α (TNF-α) levels in type 2 diabetic skin. TNF-α treatment of LECs and its specific blockade in vitro reproduced differential regulation of a gene set that led to enhanced LEC mobility and macrophage attachment, which was mediated by the LEC-derived chemokine CXCL10. This study identifies lymph vessel gene signatures directly correlated with type 2 diabetes skin manifestations. In addition, we provide evidence for paracrine cross-talk fostering macrophage recruitment to LECs as one pathophysiological process that might contribute to aberrant lymphangiogenesis and persistent inflammation in the skin.
Collapse
Affiliation(s)
- Monika Haemmerle
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Thomas Keller
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Gerda Egger
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Helga Schachner
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Carl Walter Steiner
- Department of Internal Medicine, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Dejan Stokic
- Section for Science of Complex Systems, Medical University of Vienna, Vienna, Austria
| | - Christoph Neumayer
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Markus K. Brown
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Dontscho Kerjaschki
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Brigitte Hantusch
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
- Corresponding author: Brigitte Hantusch,
| |
Collapse
|
42
|
Mirastschijski U, Martin A, Jorgensen LN, Sampson B, Ågren MS. Zinc, copper, and selenium tissue levels and their relation to subcutaneous abscess, minor surgery, and wound healing in humans. Biol Trace Elem Res 2013; 153:76-83. [PMID: 23595590 DOI: 10.1007/s12011-013-9658-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 03/27/2013] [Indexed: 11/25/2022]
Abstract
Trace element involvement in wounds left to heal by secondary intention needs clarification. We have previously reported faster healing of wounds following acute surgery compared with elective excision of pilonidal sinus disease. The effect of topical zinc on the closure of the excisional wounds was mediocre compared with placebo. In contrast, parenteral zinc, copper, and selenium combined appear effective for wound healing in humans. We have investigated zinc, copper, and selenium with respect to (a) impact of acute versus chronic pilonidal sinus and (b) regional concentrations within granulating wounds treated topically with placebo or zinc in 42 (33 males) pilonidal disease patients. Baseline serum and skin concentrations of copper correlated (r S = 0.351, p = 0.033, n = 37), but not of zinc or selenium. Patients with abscesses had elevated serum C-reactive protein (CRP) and copper levels (+29 %; p < 0.001) compared with the elective patients consistent with the strong correlation between serum copper and CRP (r S = 0.715, p < 0.0005, n = 41). Seven days after elective surgery, serum CRP and copper levels were elevated (p = 0.010) versus preoperative values. The copper concentration in wound edges was higher than in periwound skin (p < 0.0005) and wound base (p = 0.010). Selenium levels were increased in wound edge compared to wound base (p = 0.003). Topical zinc oxide treatment doubled (p < 0.050) zinc concentrations in the three tissue localizations without concomitant significant changes of copper or selenium levels. In conclusion, copper and selenium are mobilized to injured sites possibly to enhance host defense and early wound healing mechanisms that are complementary to the necessity of zinc for matrix metalloproteinase activity.
Collapse
Affiliation(s)
- Ursula Mirastschijski
- Department of Surgery K, Bispebjerg Hospital, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
43
|
Wang T, Feng Y, Sun H, Zhang L, Hao L, Shi C, Wang J, Li R, Ran X, Su Y, Zou Z. miR-21 regulates skin wound healing by targeting multiple aspects of the healing process. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 181:1911-20. [PMID: 23159215 DOI: 10.1016/j.ajpath.2012.08.022] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Revised: 08/02/2012] [Accepted: 08/15/2012] [Indexed: 01/04/2023]
Abstract
With the clarification of the important roles of microRNAs (miRNAs) in diverse physiologic and pathologic processes, the effects of miRNAs in wound healing have attracted more attention recently. However, the global pattern of miRNA expression in wound tissue is still unknown. In the present study, we depicted the miRNA profile and identified at least 54 miRNAs, including miR-21, changed for more than twofold at the stage of granulation formation during wound healing. These miRNAs were closely related to the major events of wound healing, including cell migration and proliferation, angiogenesis, and matrix remolding. Furthermore, we found that miR-21 was up-regulated after skin injury, mainly in activated and migrating epithelial cells of epidermis and mesenchymal cells of dermis. Locally antagonizing miR-21 by directly injecting antagomir to wound edge caused significant delay of wound closure with impaired collagen deposition. Unexpectedly, we found wounds treated with miR-21 antagomir had an obvious defect in wound contraction at an early stage of wound healing. The significant role of miR-21 in wound contraction was further confirmed by in vivo gain-of-function and in vitro loss-of-function experiments. In conclusion, the present study has for the first time depicted miRNA profiling of wound healing and demonstrated the involvement of miR-21 in regulating the wound contraction and collagen deposition. These results suggest that miR-21 may be a new medical target in skin wound manipulation.
Collapse
Affiliation(s)
- Tao Wang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burn and Combined Injury, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Yahaya B, McLachlan G, McCorquodale C, Collie D. Gene expression changes associated with the airway wall response to injury. PLoS One 2013; 8:e58930. [PMID: 23593124 PMCID: PMC3621906 DOI: 10.1371/journal.pone.0058930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 02/08/2013] [Indexed: 12/20/2022] Open
Abstract
Background Understanding the way in which the airway heals in response to injury is fundamental to dissecting the mechanisms underlying airway disease pathology. As only limited data is available in relation to the in vivo characterisation of the molecular features of repair in the airway we sought to characterise the dynamic changes in gene expression that are associated with the early response to physical injury in the airway wall. Methodology/Principal Findings We profiled gene expression changes in the airway wall using a large animal model of physical injury comprising bronchial brush biopsy in anaesthetised sheep. The experimental design featured sequential studies in the same animals over the course of a week and yielded data relating to the response at 6 hours, and 1, 3 and 7 days after injury. Notable features of the transcriptional response included the early and sustained preponderance of down-regulated genes associated with angiogenesis and immune cell activation, selection and differentiation. Later features of the response included the up-regulation of cell cycle genes at d1 and d3, and the latter pronounced up-regulation of extracellular matrix-related genes at d3 and d7. Conclusions/Significance It is possible to follow the airway wall response to physical injury in the same animal over the course of time. Transcriptional changes featured coordinate expression of functionally related genes in a reproducible manner both within and between animals. This characterisation will provide a foundation against which to assess the perturbations that accompany airway disease pathologies of comparative relevance.
Collapse
Affiliation(s)
- Badrul Yahaya
- Cluster for Regenerative Medicine, Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, Bandar Putra Bertam, Kepala Batas, Penang, Malaysia.
| | | | | | | |
Collapse
|
45
|
Cordeiro JV, Jacinto A. The role of transcription-independent damage signals in the initiation of epithelial wound healing. Nat Rev Mol Cell Biol 2013; 14:249-62. [PMID: 23443750 DOI: 10.1038/nrm3541] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Wound healing is an essential biological process that comprises sequential steps aimed at restoring the architecture and function of damaged cells and tissues. This process begins with conserved damage signals, such as Ca(2+), hydrogen peroxide (H2O2) and ATP, that diffuse through epithelial tissues and initiate immediate gene transcription-independent cellular effects, including cell shape changes, the formation of functional actomyosin structures and the recruitment of immune cells. These events integrate the ensuing transcription of specific wound response genes that further advance the wound healing response. The immediate importance of transcription-independent damage signals illustrates that healing a wound begins as soon as damage occurs.
Collapse
Affiliation(s)
- João V Cordeiro
- Centro de Estudos de Doenças Crónicas (CEDOC), Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria, 130, 1169-056 Lisboa, Portugal
| | | |
Collapse
|
46
|
Brubaker AL, Rendon JL, Ramirez L, Choudhry MA, Kovacs EJ. Reduced neutrophil chemotaxis and infiltration contributes to delayed resolution of cutaneous wound infection with advanced age. THE JOURNAL OF IMMUNOLOGY 2013; 190:1746-57. [PMID: 23319733 DOI: 10.4049/jimmunol.1201213] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Advanced age is associated with alterations in innate and adaptive immune responses, which contribute to an increased risk of infection in elderly patients. Coupled with this immune dysfunction, elderly patients demonstrate impaired wound healing with elevated rates of wound dehiscence and chronic wounds. To evaluate how advanced age alters the host immune response to cutaneous wound infection, we developed a murine model of cutaneous Staphylococcus aureus wound infection in young (3-4 mo) and aged (18-20 mo) BALB/c mice. Aged mice exhibit increased bacterial colonization and delayed wound closure over time compared with young mice. These differences were not attributed to alterations in wound neutrophil or macrophage TLR2 or FcγRIII expression, or age-related changes in phagocytic potential and bactericidal activity. To evaluate the role of chemotaxis in our model, we first examined in vivo chemotaxis in the absence of wound injury to KC, a neutrophil chemokine. In response to a s.c. injection of KC, aged mice recruited fewer neutrophils at increasing doses of KC compared with young mice. This paralleled our model of wound infection, where diminished neutrophil and macrophage recruitment was observed in aged mice relative to young mice despite equivalent levels of KC, MIP-2, and MCP-1 chemokine levels at the wound site. This reduced leukocyte accumulation was also associated with lower levels of ICAM-1 in wounds from aged mice at early time points. These age-mediated defects in early neutrophil recruitment may alter the dynamics of the inflammatory phase of wound healing, impacting macrophage recruitment, bacterial clearance, and wound closure.
Collapse
Affiliation(s)
- Aleah L Brubaker
- Burn and Shock Trauma Institute, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
| | | | | | | | | |
Collapse
|
47
|
Kerkhoff C, Voss A, Scholzen TE, Averill MM, Zänker KS, Bornfeldt KE. Novel insights into the role of S100A8/A9 in skin biology. Exp Dermatol 2012; 21:822-6. [PMID: 22882537 DOI: 10.1111/j.1600-0625.2012.01571.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2012] [Indexed: 12/28/2022]
Abstract
S100A8 and S100A9 belong to the damage-associated molecular pattern molecules. They are upregulated in a number of inflammatory skin disorders. Owing to their abundance in myeloid cells, the main function of S100A8/A9 has been attributed to their role in inflammatory cells. However, it is becoming increasingly clear that they also exert important roles in epithelial cells. In this review, we discuss the context-dependent function of S100A8/A9 in epithelial cells and their impact on wound healing, psoriasis and other skin diseases.
Collapse
Affiliation(s)
- Claus Kerkhoff
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Immunology, AG EXIM, Rostock, Germany.
| | | | | | | | | | | |
Collapse
|
48
|
Toriseva M, Laato M, Carpén O, Ruohonen ST, Savontaus E, Inada M, Krane SM, Kähäri VM. MMP-13 regulates growth of wound granulation tissue and modulates gene expression signatures involved in inflammation, proteolysis, and cell viability. PLoS One 2012; 7:e42596. [PMID: 22880047 PMCID: PMC3413640 DOI: 10.1371/journal.pone.0042596] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 07/09/2012] [Indexed: 01/01/2023] Open
Abstract
Proteinases play a pivotal role in wound healing by regulating cell-matrix interactions and availability of bioactive molecules. The role of matrix metalloproteinase-13 (MMP-13) in granulation tissue growth was studied in subcutaneously implanted viscose cellulose sponge in MMP-13 knockout (Mmp13(-/-)) and wild type (WT) mice. The tissue samples were harvested at time points day 7, 14 and 21 and subjected to histological analysis and gene expression profiling. Granulation tissue growth was significantly reduced (42%) at day 21 in Mmp13(-/-) mice. Granulation tissue in Mmp13(-/-) mice showed delayed organization of myofibroblasts, increased microvascular density at day 14, and virtual absence of large vessels at day 21. Gene expression profiling identified differentially expressed genes in Mmp13(-/-) mouse granulation tissue involved in biological functions including inflammatory response, angiogenesis, cellular movement, cellular growth and proliferation and proteolysis. Among genes linked to angiogenesis, Adamts4 and Npy were significantly upregulated in early granulation tissue in Mmp13(-/-) mice, and a set of genes involved in leukocyte motility including Il6 were systematically downregulated at day 14. The expression of Pdgfd was downregulated in Mmp13(-/-) granulation tissue in all time points. The expression of matrix metalloproteinases Mmp2, Mmp3, Mmp9 was also significantly downregulated in granulation tissue of Mmp13(-/-) mice compared to WT mice. Mmp13(-/-) mouse skin fibroblasts displayed altered cell morphology and impaired ability to contract collagen gel and decreased production of MMP-2. These results provide evidence for an important role for MMP-13 in wound healing by coordinating cellular activities important in the growth and maturation of granulation tissue, including myofibroblast function, inflammation, angiogenesis, and proteolysis.
Collapse
Affiliation(s)
- Mervi Toriseva
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Matti Laato
- Department of Surgery, Turku University Hospital, Turku, Finland
| | - Olli Carpén
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland
| | - Suvi T. Ruohonen
- Department of Pharmacology, Drug Development and Therapeutics, University of Turku, Turku, Finland
| | - Eriika Savontaus
- Department of Pharmacology, Drug Development and Therapeutics, University of Turku, Turku, Finland
| | - Masaki Inada
- Department of Life Science and Biotechnology, Faculty of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Stephen M. Krane
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Veli-Matti Kähäri
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
| |
Collapse
|
49
|
Dickerson R, Deshpande B, Gnyawali U, Lynch D, Gordillo GM, Schuster D, Osei K, Roy S. Correction of aberrant NADPH oxidase activity in blood-derived mononuclear cells from type II diabetes mellitus patients by a naturally fermented papaya preparation. Antioxid Redox Signal 2012; 17:485-91. [PMID: 22369197 PMCID: PMC3365356 DOI: 10.1089/ars.2011.4292] [Citation(s) in RCA: 17] [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: 09/20/2011] [Accepted: 02/08/2012] [Indexed: 01/01/2023]
Abstract
UNLABELLED Supplementation of standardized fermented papaya preparation (FPP) to adult diabetic mice improves dermal wound healing outcomes. Peripheral blood mononuclear cells (PBMC) from type II diabetes mellitus (T2DM) patients elicit a compromised respiratory burst activity resulting in increased risk of infections for the diabetic patients. AIMS The objectives of the current study were to determine the effect of FPP supplementation on human diabetic PBMC respiratory burst activity and to understand underlying mechanisms of such action of FPP. RESULTS When stimulated with phorbol 12-myristate 13-acetate, the production of reactive oxygen species by T2DM PBMC was markedly compromised compared to that of the PBMC from non-DM donors. FPP treated ex vivo improved respiratory burst outcomes in T2DM PBMC. FPP treatment significantly increased phosphorylation of the p47phox subunit of NADPH oxidase. In addition, the protein and mRNA expression of Rac2 was potently upregulated after FPP supplemention. The proximal human Rac2 gene promoter is G-C rich and contains consensus binding sites for Sp1 and AP-1. While FPP had no significant effect on the AP-1 DNA binding activity, the Sp1 DNA binding activity was significantly upregulated in PBMC after treatment of the cells with FPP. INNOVATION This work provided first evidence that compromised respiratory burst performance of T2DM PBMC may be corrected by a nutritional supplement. CONCLUSION FPP can correct respiratory burst performance of T2DM PBMC via an Sp-1-dependant pathway. Studies testing the outcome of FPP supplementation in diabetic patients are warranted.
Collapse
Affiliation(s)
- Ryan Dickerson
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
| | - Bhakthi Deshpande
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
| | - Urmila Gnyawali
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
| | - Debbie Lynch
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
| | - Gayle M. Gordillo
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
| | - Dara Schuster
- Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Kwame Osei
- Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Sashwati Roy
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, Ohio
- Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, Ohio
| |
Collapse
|
50
|
Ganesh K, Das A, Dickerson R, Khanna S, Parinandi NL, Gordillo GM, Sen CK, Roy S. Prostaglandin E₂ induces oncostatin M expression in human chronic wound macrophages through Axl receptor tyrosine kinase pathway. THE JOURNAL OF IMMUNOLOGY 2012; 189:2563-73. [PMID: 22844123 DOI: 10.4049/jimmunol.1102762] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Monocytes and macrophages (m) are plastic cells whose functions are governed by microenvironmental cues. Wound fluid bathing the wound tissue reflects the wound microenvironment. Current literature on wound inflammation is primarily based on the study of blood monocyte-derived macrophages, cells that have never been exposed to the wound microenvironment. We sought to compare pair-matched monocyte-derived macrophages with m isolated from chronic wounds of patients. Oncostatin M (OSM) was differentially overexpressed in pair-matched wound m. Both PGE₂ and its metabolite 13,14-dihydro-15-keto-PGE₂ (PGE-M) were abundant in wound fluid and induced OSM in wound-site m. Consistently, induction of OSM mRNA was observed in m isolated from PGE₂-enriched polyvinyl alcohol sponges implanted in murine wounds. Treatment of human THP-1 cell-derived m with PGE₂ or PGE-M caused dose-dependent induction of OSM. Characterization of the signal transduction pathways demonstrated the involvement of EP4 receptor and cAMP signaling. In human m, PGE₂ phosphorylated Axl, a receptor tyrosine kinase (RTK). Axl phosphorylation was also induced by a cAMP analogue demonstrating interplay between the cAMP and RTK pathways. PGE₂-dependent Axl phosphorylation led to AP-1 transactivation, which is directly implicated in inducible expression of OSM. Treatment of human m or mice excisional wounds with recombinant OSM resulted in an anti-inflammatory response as manifested by attenuated expression of endotoxin-induced TNF-α and IL-1β. OSM treatment also improved wound closure during the early inflammatory phase of healing. In summary, this work recognizes PGE₂ in the wound fluid as a potent inducer of m OSM, a cytokine with an anti-inflammatory role in cutaneous wound healing.
Collapse
Affiliation(s)
- Kasturi Ganesh
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|