201
|
Overview and Compartmentalization of the Immune System. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00019-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
202
|
Wei F, Xiao Y. Modulation of the Osteoimmune Environment in the Development of Biomaterials for Osteogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1077:69-86. [DOI: 10.1007/978-981-13-0947-2_5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
203
|
Janner SFM, Gahlert M, Bosshardt DD, Roehling S, Milz S, Higginbottom F, Buser D, Cochran DL. Bone response to functionally loaded, two-piece zirconia implants: A preclinical histometric study. Clin Oral Implants Res 2017; 29:277-289. [PMID: 29288505 DOI: 10.1111/clr.13112] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2017] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To evaluate the bone response to a two-piece zirconia implant in comparison with a control titanium implant in the canine mandible 4 and 16 weeks after restoration. MATERIAL AND METHODS Zirconia and titanium implants were alternately placed bilaterally in healed mandibular molar and premolar sites of five canines. Full-ceramic single-tooth restorations were cemented after 6 weeks of transmucosal healing, allowing for full functional loading of the implants. Histologic and histometric analyses were performed on orofacial and mesiodistal undecalcified sections of the specimens obtained upon sacrifice after 4 and 16 weeks of functional loading. Bone-to-implant contact (BIC), multinucleated giant cells-to-implant contact (MIC), crestal bone level, and peri-implant bone density were histometrically assessed. RESULTS All 60 implants and 60 restorations were still in function after 4 and 16 weeks of loading in both test and control groups. No implant loss, no implant or abutment fracture, and no chipping of the restorations could be detected. Histometric analysis showed no statistically significant differences between zirconia and titanium implants in BIC, crestal bone level, and peri-implant bone density at both time points. Between 4 and 16 weeks, the crestal bone level around zirconia implants showed a small but statistically significant increase in its distance from the implant shoulder. MIC was very low on both implant types and both time points and decreased statistically significantly overtime. CONCLUSION The present two-piece zirconia implant showed a similar bone integration compared to the titanium implant with similar surface morphology after 4 and 16 weeks of loading.
Collapse
Affiliation(s)
- Simone F M Janner
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Michael Gahlert
- Clinic for Oral and Cranio-Maxillofacial Surgery, Hightech Research Center, University Hospital Basel, University of Basel, Basel, Switzerland.,Private Practice, Munich, Germany
| | - Dieter D Bosshardt
- Robert K. Schenk Laboratory of Oral Histology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Stefan Roehling
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Clinic for Oral and Cranio-Maxillofacial Surgery, Hightech Research Center, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stefan Milz
- Anatomische Anstalt, Ludwig-Maximilians University, Munich, Germany
| | | | - Daniel Buser
- Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - David L Cochran
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
204
|
Thulabandu V, Chen D, Atit RP. Dermal fibroblast in cutaneous development and healing. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2017; 7. [PMID: 29244903 DOI: 10.1002/wdev.307] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 09/27/2017] [Accepted: 10/07/2017] [Indexed: 01/09/2023]
Abstract
The skin is the largest organ of the body and is composed of two layers: the overlying epidermis and the underlying dermis. The dermal fibroblasts originate from distinct locations of the embryo and contain the positional identity and patterning information in the skin. The dermal fibroblast progenitors differentiate into various cell types that are fated to perform specific functions such as hair follicle initiation and scar formation during wound healing. Recent studies have revealed the heterogeneity and plasticity of dermal fibroblasts within skin, which has implications for skin disease and tissue engineering. The objective of this review is to frame our current understanding and provide new insights on the origin and differentiation of dermal fibroblasts and their function during cutaneous development and healing. WIREs Dev Biol 2018, 7:e307. doi: 10.1002/wdev.307 This article is categorized under: Birth Defects > Organ Anomalies Signaling Pathways > Cell Fate Signaling Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Venkata Thulabandu
- Department of Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Demeng Chen
- Department of Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Radhika P Atit
- Department of Biology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
205
|
Redox regulation of leukocyte-derived microparticle release and protein content in response to cold physical plasma-derived oxidants. CLINICAL PLASMA MEDICINE 2017. [DOI: 10.1016/j.cpme.2017.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
206
|
Sun G. Pro-Regenerative Hydrogel Restores Scarless Skin during Cutaneous Wound Healing. Adv Healthc Mater 2017; 6. [PMID: 28945013 DOI: 10.1002/adhm.201700659] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 08/09/2017] [Indexed: 11/06/2022]
Abstract
The transformation of fibrotic healing process to regenerative one has great potential to fully restore wounded skin. The M2 macrophage phenotype promotes constructive tissue remodeling and instructs tissue repair in a regenerative manner. It is hypothesized that hydrogels that can establish robustness of endogenous cells to regulate M2 phenotype will promote constructive dermal remodeling. Toward this end, a series of dextran-based bioabsorbable hydrogels are developed and self-crosslinkable dextran-isocyanatoethyl methacrylate-ethylamine (DexIEME) is identified as the potential scaffold. The initial screening study revealed that DexIEME has superior biocompatibility in varying concentrations. Although DexIEME brings about low proinflammatory responses, it promotes M2 macrophage phenotype. Then the optimized hydrogel formulation is tested for acute skin injuries using both murine and porcine models. Preliminary data demonstrated that the innovative DexIEME hydrogel promotes complete skin regeneration with hair regrowth on pre-existing scars, while untreated scars remain intact. Preclinical studies further demonstrated that the DexIEME hydrogel regenerated perfect skin during deep porcine wound healing. Overall, the approach to investigate immune-modulated hydrogels yields pro-regenerative DexIEME hydrogel, which may lead to greater clinical success in treating deep dermal injury and attenuating scar formation.
Collapse
Affiliation(s)
- Guoming Sun
- Sunogel Biotechnologies Inc.; 9 W Ridgely Road Ste 270 Lutherville Timonium MD 21093 USA
| |
Collapse
|
207
|
Amici SA, Dong J, Guerau-de-Arellano M. Molecular Mechanisms Modulating the Phenotype of Macrophages and Microglia. Front Immunol 2017; 8:1520. [PMID: 29176977 PMCID: PMC5686097 DOI: 10.3389/fimmu.2017.01520] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/26/2017] [Indexed: 12/24/2022] Open
Abstract
Macrophages and microglia play crucial roles during central nervous system development, homeostasis and acute events such as infection or injury. The diverse functions of tissue macrophages and microglia are mirrored by equally diverse phenotypes. A model of inflammatory/M1 versus a resolution phase/M2 macrophages has been widely used. However, the complexity of macrophage function can only be achieved by the existence of varied, plastic and tridimensional macrophage phenotypes. Understanding how tissue macrophages integrate environmental signals via molecular programs to define pathogen/injury inflammatory responses provides an opportunity to better understand the multilayered nature of macrophages, as well as target and modulate cellular programs to control excessive inflammation. This is particularly important in MS and other neuroinflammatory diseases, where chronic inflammatory macrophage and microglial responses may contribute to pathology. Here, we perform a comprehensive review of our current understanding of how molecular pathways modulate tissue macrophage phenotype, covering both classic pathways and the emerging role of microRNAs, receptor-tyrosine kinases and metabolism in macrophage phenotype. In addition, we discuss pathway parallels in microglia, novel markers helpful in the identification of peripheral macrophages versus microglia and markers linked to their phenotype.
Collapse
Affiliation(s)
- Stephanie A Amici
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Joycelyn Dong
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States.,McCormick School of Engineering, Division of Biomedical Engineering, Northwestern University, Evanston, IL, United States
| | - Mireia Guerau-de-Arellano
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States.,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States.,Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States.,Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
208
|
Makrantonaki E, Wlaschek M, Scharffetter-Kochanek K. Pathogenesis of wound healing disorders in the elderly. J Dtsch Dermatol Ges 2017; 15:255-275. [PMID: 28252848 DOI: 10.1111/ddg.13199] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/21/2016] [Indexed: 12/25/2022]
Abstract
The elderly constitute the age group most susceptible to wound healing disorders and chronic wounds, the most prevalent being venous leg ulcers, pressure ulcers, and diabetic foot ulcers. However, other age-associated diseases should also be taken into consideration in the diagnostic workup of chronic wounds, and not be underestimated. A better understanding of the pathomechanisms involved in the wound healing process is of key importance in combatting the difficulties associated with the treatment of chronic wounds. In recent decades, considerable progress has been made in the development of pioneering therapeutic strategies for chronic wounds. In this context, the use of growth factors and cytokines, tissue engineering, and cell therapy - including stem cells - have proven very promising. Nevertheless, prior to their introduction into routine clinical practice, large controlled clinical trials are required to assess the safety of these techniques.
Collapse
Affiliation(s)
| | - Meinhard Wlaschek
- Department of Dermatology and Allergic Diseases, University Medical Center Ulm
| | | |
Collapse
|
209
|
He R, Yin H, Yuan B, Liu T, Luo L, Huang P, Dai L, Zeng K. IL-33 improves wound healing through enhanced M2 macrophage polarization in diabetic mice. Mol Immunol 2017; 90:42-49. [PMID: 28697404 DOI: 10.1016/j.molimm.2017.06.249] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/26/2017] [Accepted: 06/30/2017] [Indexed: 12/22/2022]
Abstract
IL-33 is a newly discovered member of the IL-1 family and has been identified as a potent inducer of Th2 type immunity. Emerging evidence imply that IL-33 may also act as an alarm to alert the immune system when released by epithelial barrier tissues during trauma or infection. In this study, we further investigate the potential efficacy of IL-33 on dermal wound healing in streptozotocin-induced diabetic mice. A full-thickness skin wound was generated on the back of diabetic mice and treated with IL-33 or vehicle topically. Our data showed that IL-33 delivery contributed to diabetic wound closure with wounds gaping narrower and exhibiting elevated re-epithelialization. IL-33 promoted the new extracellular matrix (ECM) deposition and angiogenesis formation, which indicates an important role of IL-33 on matrix synthesis and neovascularization. Meanwhile, IL-33 accelerated the development of M2 macrophages in wound sites in vivo, and amplified IL-13-induced polarization of bone marrow-derived macrophages toward a M2 phenotype in vitro. Furthermore, IL-33-amplified M2 macrophages augmented the proliferation of fibroblasts and ECM deposition. All together, these results strongly suggest manipulation of IL-33-mediated signal might be a potential therapeutic approach for diabetic skin wounds.
Collapse
Affiliation(s)
- Rongguo He
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Dermatology, Guangzhou Twelfth People's Hospital, Guangzhou 510620, China
| | - Hui Yin
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Baohong Yuan
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Tao Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Li Luo
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ping Huang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Liangcheng Dai
- Intensive Care Unit, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Kang Zeng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
210
|
Yuan X, Cao H, Wang J, Tang K, Li B, Zhao Y, Cheng M, Qin H, Liu X, Zhang X. Immunomodulatory Effects of Calcium and Strontium Co-Doped Titanium Oxides on Osteogenesis. Front Immunol 2017; 8:1196. [PMID: 29033930 PMCID: PMC5626827 DOI: 10.3389/fimmu.2017.01196] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/11/2017] [Indexed: 01/07/2023] Open
Abstract
The effects of calcium (Ca) or strontium (Sr) on host osteogenesis and immune responses have been investigated separately. In clinical practice, these two elements may both be present around an orthopedic device, but their potential synergistic effects on osteogenesis and the immune response have not been explored to date. In this work, we investigated the immunomodulatory effects of Ca and Sr co-doped titanium oxides on osteogenesis in vitro using the mouse macrophage cell line RAW 264.7 alone and in co-culture with mouse bone mesenchymal stem cells (BMSCs), and in vivo using a mouse air-pouch model. Coatings containing Ca and Sr at different concentration ratios were fabricated on titanium substrates using micro-arc oxidation and electrochemical treatment. The in vitro and in vivo results demonstrated that the Ca and Sr concentration ratio has a marked influence on macrophage polarization. The coating with a Ca/Sr ratio of 2:1 was superior to those with other Ca and/or Sr concentrations in terms of modulating M2 polarization, which enhanced osteogenic differentiation of mouse BMSCs in co-culture. These findings suggest that the osteoimmunomodulatory effect of a titanium-oxide coating can be enhanced by modulating the concentration ratio of its components.
Collapse
Affiliation(s)
- Xiangwei Yuan
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Huiliang Cao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Jiaxing Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Kaiwei Tang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Bin Li
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yaochao Zhao
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Mengqi Cheng
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Qin
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Xianlong Zhang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
211
|
Yu B, Alboslemy T, Safadi F, Kim MH. Glycoprotein Nonmelanoma Clone B Regulates the Crosstalk between Macrophages and Mesenchymal Stem Cells toward Wound Repair. J Invest Dermatol 2017; 138:219-227. [PMID: 28899684 DOI: 10.1016/j.jid.2017.08.034] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 08/03/2017] [Accepted: 08/08/2017] [Indexed: 11/28/2022]
Abstract
The process of wound repair requires the coordinated participation of multiple types of cells, which are sequentially recruited during the healing process. In response to tissue injury, both macrophages and mesenchymal stem cells (MSCs) are recruited to the site of injury, where they participate in the repair process. Despite considerable understanding of the role of each cell type in the process of wound repair, the nature of the dynamic interplay between these two cell types and how this interaction influences the process of wound repair are not well understood. Here, using an in vivo model of cutaneous wound healing in mice, we provide evidence that GPNMB is functionally important in promoting the recruitment of MSCs to the site of skin injury, which in turn modulates inflammatory responses by directing the M2 polarization of macrophages in acute wound healing. Furthermore, we show that GPNMB activity is impaired in a diabetic wound environment, which is associated with impaired MSC recruitment that is reversed by the topical administration of recombinant GPNMB protein to the wounds of diabetic mice. Our study provides important insight into the crosstalk between macrophages and endogenous MSCs toward wound repair.
Collapse
Affiliation(s)
- Bing Yu
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Talib Alboslemy
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
| | - Fayez Safadi
- Department of Neurobiology and Anatomy, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Min-Ho Kim
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA; School of Biomedical Sciences, Kent State University, Kent, Ohio, USA.
| |
Collapse
|
212
|
Rotty JD, Brighton HE, Craig SL, Asokan SB, Cheng N, Ting JP, Bear JE. Arp2/3 Complex Is Required for Macrophage Integrin Functions but Is Dispensable for FcR Phagocytosis and In Vivo Motility. Dev Cell 2017; 42:498-513.e6. [PMID: 28867487 DOI: 10.1016/j.devcel.2017.08.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 06/19/2017] [Accepted: 08/02/2017] [Indexed: 11/18/2022]
Abstract
The Arp2/3 complex nucleates branched actin, forming networks involved in lamellipodial protrusion, phagocytosis, and cell adhesion. We derived primary bone marrow macrophages lacking Arp2/3 complex (Arpc2-/-) and directly tested its role in macrophage functions. Despite protrusion and actin assembly defects, Arpc2-/- macrophages competently phagocytose via FcR and chemotax toward CSF and CX3CL1. However, CR3 phagocytosis and fibronectin haptotaxis, both integrin-dependent processes, are disrupted. Integrin-responsive actin assembly and αM/β2 integrin localization are compromised in Arpc2-/- cells. Using an in vivo system to observe endogenous monocytes migrating toward full-thickness ear wounds we found that Arpc2-/- monocytes maintain cell speeds and directionality similar to control. Our work reveals that the Arp2/3 complex is not a general requirement for phagocytosis or chemotaxis but is a critical driver of integrin-dependent processes. We demonstrate further that cells lacking Arp2/3 complex function in vivo remain capable of executing important physiological responses that require rapid directional motility.
Collapse
Affiliation(s)
- Jeremy D Rotty
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hailey E Brighton
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Stephanie L Craig
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sreeja B Asokan
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ning Cheng
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Oral Biology Curriculum, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jenny P Ting
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Oral Biology Curriculum, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - James E Bear
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
213
|
Transcriptional mechanisms that control expression of the macrophage colony-stimulating factor receptor locus. Clin Sci (Lond) 2017; 131:2161-2182. [DOI: 10.1042/cs20170238] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/22/2017] [Accepted: 06/11/2017] [Indexed: 12/17/2022]
Abstract
The proliferation, differentiation, and survival of cells of the macrophage lineage depends upon signals from the macrophage colony-stimulating factor (CSF) receptor (CSF1R). CSF1R is expressed by embryonic macrophages and induced early in adult hematopoiesis, upon commitment of multipotent progenitors to the myeloid lineage. Transcriptional activation of CSF1R requires interaction between members of the E26 transformation-specific family of transcription factors (Ets) (notably PU.1), C/EBP, RUNX, AP-1/ATF, interferon regulatory factor (IRF), STAT, KLF, REL, FUS/TLS (fused in sarcoma/ranslocated in liposarcoma) families, and conserved regulatory elements within the mouse and human CSF1R locus. One element, the Fms-intronic regulatory element (FIRE), within intron 2, is conserved functionally across all the amniotes. Lineage commitment in multipotent progenitors also requires down-regulation of specific transcription factors such as MYB, FLI1, basic leucine zipper transcriptional factor ATF-like (BATF3), GATA-1, and PAX5 that contribute to differentiation of alternative lineages and repress CSF1R transcription. Many of these transcription factors regulate each other, interact at the protein level, and are themselves downstream targets of CSF1R signaling. Control of CSF1R transcription involves feed–forward and feedback signaling in which CSF1R is both a target and a participant; and dysregulation of CSF1R expression and/or function is associated with numerous pathological conditions. In this review, we describe the regulatory network behind CSF1R expression during differentiation and development of cells of the mononuclear phagocyte system.
Collapse
|
214
|
Gaffney L, Wrona EA, Freytes DO. Potential Synergistic Effects of Stem Cells and Extracellular Matrix Scaffolds. ACS Biomater Sci Eng 2017. [DOI: 10.1021/acsbiomaterials.7b00083] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Lewis Gaffney
- Joint Department of Biomedical Engineering, North Carolina State University/University of North Carolina-Chapel Hill, Raleigh, North Carolina 27695, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Emily A. Wrona
- Joint Department of Biomedical Engineering, North Carolina State University/University of North Carolina-Chapel Hill, Raleigh, North Carolina 27695, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Donald O. Freytes
- Joint Department of Biomedical Engineering, North Carolina State University/University of North Carolina-Chapel Hill, Raleigh, North Carolina 27695, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| |
Collapse
|
215
|
Hesketh M, Sahin KB, West ZE, Murray RZ. Macrophage Phenotypes Regulate Scar Formation and Chronic Wound Healing. Int J Mol Sci 2017; 18:ijms18071545. [PMID: 28714933 PMCID: PMC5536033 DOI: 10.3390/ijms18071545] [Citation(s) in RCA: 559] [Impact Index Per Article: 69.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 07/13/2017] [Accepted: 07/16/2017] [Indexed: 12/21/2022] Open
Abstract
Macrophages and inflammation play a beneficial role during wound repair with macrophages regulating a wide range of processes, such as removal of dead cells, debris and pathogens, through to extracellular matrix deposition re-vascularisation and wound re-epithelialisation. To perform this range of functions, these cells develop distinct phenotypes over the course of wound healing. They can present with a pro-inflammatory M1 phenotype, more often found in the early stages of repair, through to anti-inflammatory M2 phenotypes that are pro-repair in the latter stages of wound healing. There is a continuum of phenotypes between these ranges with some cells sharing phenotypes of both M1 and M2 macrophages. One of the less pleasant consequences of quick closure, namely the replacement with scar tissue, is also regulated by macrophages, through their promotion of fibroblast proliferation, myofibroblast differentiation and collagen deposition. Alterations in macrophage number and phenotype disrupt this process and can dictate the level of scar formation. It is also clear that dysregulated inflammation and altered macrophage phenotypes are responsible for hindering closure of chronic wounds. The review will discuss our current knowledge of macrophage phenotype on the repair process and how alterations in the phenotypes might alter wound closure and the final repair quality.
Collapse
Affiliation(s)
- Mark Hesketh
- The Institute for Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane QLD 4059, Australia.
| | - Katherine B Sahin
- The Institute for Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane QLD 4059, Australia.
| | - Zoe E West
- The Institute for Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane QLD 4059, Australia.
| | - Rachael Z Murray
- The Institute for Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane QLD 4059, Australia.
| |
Collapse
|
216
|
D’asta F, Halstead F, Harrison P, Zecchi Orlandini S, Moiemen N, Lord J. The contribution of leucocytes to the antimicrobial activity of platelet-rich plasma preparations: A systematic review. Platelets 2017; 29:9-20. [DOI: 10.1080/09537104.2017.1317731] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Federica D’asta
- Birmingham Children’s Hospital, Burns Centre, Steelhouse Lane, Birmingham, UK
| | - Fenella Halstead
- NIHR Surgical Reconstruction and Microbiology Research, CentreQueen Elizabeth Hospital, Birmingham, UK
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Paul Harrison
- NIHR Surgical Reconstruction and Microbiology Research, CentreQueen Elizabeth Hospital, Birmingham, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Sandra Zecchi Orlandini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Naiem Moiemen
- NIHR Surgical Reconstruction and Microbiology Research, CentreQueen Elizabeth Hospital, Birmingham, UK
- The Scar Free Foundation Centre for Burns Research, Queen Elizabeth Hospital, Birmingham, UK
| | - Janet Lord
- NIHR Surgical Reconstruction and Microbiology Research, CentreQueen Elizabeth Hospital, Birmingham, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
217
|
Erndt-Marino JD, Jimenez-Vergara AC, Diaz-Rodriguez P, Kulwatno J, Diaz-Quiroz JF, Thibeault S, Hahn MS. In vitro evaluation of a basic fibroblast growth factor-containing hydrogel toward vocal fold lamina propria scar treatment. J Biomed Mater Res B Appl Biomater 2017; 106:1258-1267. [PMID: 28580765 DOI: 10.1002/jbm.b.33936] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 04/11/2017] [Accepted: 05/16/2017] [Indexed: 12/25/2022]
Abstract
Scarring of the vocal fold lamina propria can lead to debilitating voice disorders that can significantly impair quality of life. The reduced pliability of the scar tissue-which diminishes proper vocal fold vibratory efficiency-results in part from abnormal extracellular matrix (ECM) deposition by vocal fold fibroblasts (VFF) that have taken on a fibrotic phenotype. To address this issue, bioactive materials containing cytokines and/or growth factors may provide a platform to transition fibrotic VFF within the scarred tissue toward an anti-fibrotic phenotype, thereby improving the quality of ECM within the scar tissue. However, for such an approach to be most effective, the acute host response resulting from biomaterial insertion/injection likely also needs to be considered. The goal of the present work was to evaluate the anti-fibrotic and anti-inflammatory capacity of an injectable hydrogel containing tethered basic fibroblast growth factor (bFGF) in the dual context of scar and biomaterial-induced acute inflammation. An in vitro co-culture system was utilized containing both activated, fibrotic VFF and activated, pro-inflammatory macrophages (MΦ) within a 3D poly(ethylene glycol) diacrylate (PEGDA) hydrogel containing tethered bFGF. Following 72 h of culture, alterations in VFF and macrophage phenotype were evaluated relative to mono-culture and co-culture controls. In our co-culture system, bFGF reduced the production of fibrotic markers collagen type I, α smooth muscle actin, and biglycan by activated VFF and promoted wound-healing/anti-inflammatory marker expression in activated MΦ. Cumulatively, these data indicate that bFGF-containing hydrogels warrant further investigation for the treatment of vocal fold lamina propria scar. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1258-1267, 2018.
Collapse
Affiliation(s)
- Josh D Erndt-Marino
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | | | | | - Jonathan Kulwatno
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | | | - Susan Thibeault
- Department of Surgery, Division of Otolaryngology-Head & Neck Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Mariah S Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
218
|
De Schryver M, Leemans A, Pintelon I, Cappoen D, Maes L, Caljon G, Cos P, Delputte PL. Comparative analysis of the internalization of the macrophage receptor sialoadhesin in human and mouse primary macrophages and cell lines. Immunobiology 2017; 222:797-806. [DOI: 10.1016/j.imbio.2016.11.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 11/18/2016] [Accepted: 11/20/2016] [Indexed: 01/05/2023]
|
219
|
Nishikai-Yan Shen T, Kanazawa S, Kado M, Okada K, Luo L, Hayashi A, Mizuno H, Tanaka R. Interleukin-6 stimulates Akt and p38 MAPK phosphorylation and fibroblast migration in non-diabetic but not diabetic mice. PLoS One 2017; 12:e0178232. [PMID: 28542434 PMCID: PMC5441644 DOI: 10.1371/journal.pone.0178232] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 05/03/2017] [Indexed: 12/12/2022] Open
Abstract
Persistent inflammatory environment and abnormal macrophage activation are characteristics of chronic diabetic wounds. Here, we attempted to characterize the differences in macrophage activation and temporal variations in cytokine expression in diabetic and non-diabetic wounds, with a focus on interleukin (IL)-6 mRNA expression and the p38 MAPK and PI3K/Akt signaling pathways. Cutaneous wound closure, CD68- and arginase-1 (Arg-1)-expressing macrophages, and cytokine mRNA expression were examined in non-diabetic and streptozotocin-induced type 1 diabetic mice at different time points after injury. The effect of IL-6 on p38 MAPK and Akt phosphorylation was investigated, and an in vitro scratch assay was performed to determine the role of IL-6 in primary skin fibroblast migration. Before injury, mRNA expression levels of the inflammatory markers iNOS, IL-6, and TNF-α were higher in diabetic mice; however, IL-6 expression was significantly lower 6 h post injury in diabetic wounds than that in non-diabetic wounds. Non-diabetic wounds exhibited increased p38 MAPK and Akt phosphorylation; however, no such increase was found in diabetic wounds. In fibroblasts from non-diabetic mice, IL-6 increased the phosphorylation of p38 MAPK and levels of its downstream factor CREB, and also significantly increased Akt phosphorylation and levels of its upstream factor P13K. These effects of IL-6 were not detected in fibroblasts derived from the diabetic mice. In scratch assays, IL-6 stimulated the migration of primary cultured skin fibroblasts from the non-diabetic mice, and the inhibition of p38 MAPK was found to markedly suppress IL-6–stimulated fibroblast migration. These findings underscore the critical differences between diabetic and non-diabetic wounds in terms of macrophage activation, cytokine mRNA expression profile, and involvement of the IL-6-stimulated p38 MAPK–Akt signaling pathway. Aberrant macrophage activation and abnormalities in the cytokine mRNA expression profile during different phases of wound healing should be addressed when designing effective therapeutic modalities for refractory diabetic wounds.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Cell Movement/drug effects
- Cell Movement/physiology
- Cells, Cultured
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Interleukin-6/administration & dosage
- Interleukin-6/metabolism
- Macrophages/drug effects
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Mice, Inbred C57BL
- Phosphorylation/drug effects
- Phosphorylation/physiology
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Messenger/metabolism
- Skin/drug effects
- Skin/metabolism
- Skin/pathology
- Wound Healing/drug effects
- Wound Healing/physiology
- p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Tsubame Nishikai-Yan Shen
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Shigeyuki Kanazawa
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
- Department of Plastic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Makiko Kado
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Kayoko Okada
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Lin Luo
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Ayato Hayashi
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiroshi Mizuno
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Rica Tanaka
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
- * E-mail:
| |
Collapse
|
220
|
Smith TD, Nagalla RR, Chen EY, Liu WF. Harnessing macrophage plasticity for tissue regeneration. Adv Drug Deliv Rev 2017; 114:193-205. [PMID: 28449872 DOI: 10.1016/j.addr.2017.04.012] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/19/2017] [Accepted: 04/21/2017] [Indexed: 12/25/2022]
Abstract
Macrophages are versatile and plastic effector cells of the immune system, and contribute to diverse immune functions including pathogen or apoptotic cell removal, inflammatory activation and resolution, and tissue healing. Macrophages function as signaling regulators and amplifiers, and influencing their activity is a powerful approach for controlling inflammation or inducing a wound-healing response in regenerative medicine. This review discusses biomaterials-based approaches for altering macrophage activity, approaches for targeting drugs to macrophages, and approaches for delivering macrophages themselves as a therapeutic intervention.
Collapse
|
221
|
Cancedda R, Bollini S, Descalzi F, Mastrogiacomo M, Tasso R. Learning from Mother Nature: Innovative Tools to Boost Endogenous Repair of Critical or Difficult-to-Heal Large Tissue Defects. Front Bioeng Biotechnol 2017; 5:28. [PMID: 28503549 PMCID: PMC5408079 DOI: 10.3389/fbioe.2017.00028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/10/2017] [Indexed: 12/16/2022] Open
Abstract
For repair of chronic or difficult-to-heal tissue lesions and defects, major constraints exist to a broad application of cell therapy and tissue engineering approaches, i.e., transplantation of “ex vivo” expanded autologous stem/progenitor cells, alone or associated with carrier biomaterials. To enable a large number of patients to benefit, new strategies should be considered. One of the main goals of contemporary regenerative medicine is to develop new regenerative therapies, inspired from Mother Nature. In all injured tissues, when platelets are activated by tissue contact, their released factors promote innate immune cell migration to the wound site. Platelet-derived factors and factors secreted by migrating immune cells create an inflammatory microenvironment, in turn, causing the activation of angiogenesis and vasculogenesis processes. Eventually, repair or regeneration of the injured tissue occurs via paracrine signals activating, mobilizing or recruiting to the wound site cells with healing potential, such as stem cells, progenitors, or undifferentiated cells derived from the reprogramming of tissue differentiated cells. This review, largely based on our studies, discusses the identification of new tools, inspired by cellular and molecular mechanisms overseeing physiological tissue healing, that could reactivate dormant endogenous regeneration mechanisms lost during evolution and ontogenesis.
Collapse
Affiliation(s)
- Ranieri Cancedda
- Biorigen Srl, Genova, Italy.,Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Sveva Bollini
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | | | | | - Roberta Tasso
- IRCCS AOU San Martino-IST National Institute of Cancer Research, Genova, Italy
| |
Collapse
|
222
|
Laurent P, Jolivel V, Manicki P, Chiu L, Contin-Bordes C, Truchetet ME, Pradeu T. Immune-Mediated Repair: A Matter of Plasticity. Front Immunol 2017; 8:454. [PMID: 28484454 PMCID: PMC5403426 DOI: 10.3389/fimmu.2017.00454] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/04/2017] [Indexed: 12/28/2022] Open
Abstract
Though the immune system is generally defined as a system of defense, it is increasingly recognized that the immune system also plays a crucial role in tissue repair and its potential dysregulations. In this review, we explore how distinct immune cell types are involved in tissue repair and how they interact in a process that is tightly regulated both spatially and temporally. We insist on the concept of immune cell plasticity which, in recent years, has proved fundamental for the success/understanding of the repair process. Overall, the perspective presented here suggests that the immune system plays a central role in the physiological robustness of the organism, and that cell plasticity contributes to the realization of this robustness.
Collapse
Affiliation(s)
- Paôline Laurent
- ImmunoConcept, UMR5164, Immunology, CNRS, University of Bordeaux, Bordeaux, France
| | - Valérie Jolivel
- ImmunoConcept, UMR5164, Immunology, CNRS, University of Bordeaux, Bordeaux, France
| | | | - Lynn Chiu
- ImmunoConcept, UMR5164, Immunology, CNRS, University of Bordeaux, Bordeaux, France
| | - Cécile Contin-Bordes
- ImmunoConcept, UMR5164, Immunology, CNRS, University of Bordeaux, Bordeaux, France.,Immunology, CHU Bordeaux Hospital, Bordeaux, France
| | - Marie-Elise Truchetet
- ImmunoConcept, UMR5164, Immunology, CNRS, University of Bordeaux, Bordeaux, France.,Rheumatology, CHU Bordeaux Hospital, Bordeaux, France
| | - Thomas Pradeu
- ImmunoConcept, UMR5164, Immunology, CNRS, University of Bordeaux, Bordeaux, France
| |
Collapse
|
223
|
Strong AL, Neumeister MW, Levi B. Stem Cells and Tissue Engineering: Regeneration of the Skin and Its Contents. Clin Plast Surg 2017; 44:635-650. [PMID: 28576253 DOI: 10.1016/j.cps.2017.02.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In this review, the authors discuss the stages of skin wound healing, the role of stem cells in accelerating skin wound healing, and the mechanism by which these stem cells may reconstitute the skin in the context of tissue engineering.
Collapse
Affiliation(s)
- Amy L Strong
- Division of Plastic Surgery, Department of Surgery, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Michael W Neumeister
- Department of Surgery, Institute for Plastic Surgery, Southern Illinois University School of Medicine, 747 North Rutledge Street, Springfield, IL 62702, USA
| | - Benjamin Levi
- Division of Plastic Surgery, Department of Surgery, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA; Burn Wound and Regenerative Medicine Laboratory, University of Michigan, 1150 West Medical Center Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
224
|
Hu K, Olsen BR. Vascular endothelial growth factor control mechanisms in skeletal growth and repair. Dev Dyn 2017; 246:227-234. [PMID: 27750398 PMCID: PMC5354946 DOI: 10.1002/dvdy.24463] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/01/2016] [Indexed: 01/04/2023] Open
Abstract
Vascular endothelial growth factor A (VEGF) is a critical regulator of vascular development and postnatal angiogenesis and homeostasis, and it is essential for bone development and repair. Blood vessels serve both as structural templates for bone formation and they provide essential cells, growth factors and minerals needed for synthesis and mineralization, as well as turnover, of the extracellular matrix in bone. Through its regulation of angiogenesis, VEGF contributes to coupling of osteogenesis to angiogenesis, and it directly controls the differentiation and function of osteoblasts and osteoclasts. In this review, we summarize the properties of VEGF and its receptors that are relevant to bone formation and repair; the roles of VEGF during development of endochondral and membranous bones; and the contributions of VEGF to bone healing during different phases of bone repair. Finally, we discuss contributions of altered VEGF function in inherited disorders with bone defects as part of their phenotypes, and we speculate on what will be required before therapeutic strategies based on VEGF modulation can be developed for clinical use to treat patients with bone growth disorders and/or compromised bone repair. Developmental Dynamics 246:227-234, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kai Hu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Bjorn R Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| |
Collapse
|
225
|
Wang X, Chen H, Tian R, Zhang Y, Drutskaya MS, Wang C, Ge J, Fan Z, Kong D, Wang X, Cai T, Zhou Y, Wang J, Wang J, Wang S, Qin Z, Jia H, Wu Y, Liu J, Nedospasov SA, Tredget EE, Lin M, Liu J, Jiang Y, Wu Y. Macrophages induce AKT/β-catenin-dependent Lgr5 + stem cell activation and hair follicle regeneration through TNF. Nat Commun 2017; 8:14091. [PMID: 28345588 PMCID: PMC5378973 DOI: 10.1038/ncomms14091] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 11/29/2016] [Indexed: 12/11/2022] Open
Abstract
Skin stem cells can regenerate epidermal appendages; however, hair follicles (HF) lost as a result of injury are barely regenerated. Here we show that macrophages in wounds activate HF stem cells, leading to telogen-anagen transition (TAT) around the wound and de novo HF regeneration, mostly through TNF signalling. Both TNF knockout and overexpression attenuate HF neogenesis in wounds, suggesting dose-dependent induction of HF neogenesis by TNF, which is consistent with TNF-induced AKT signalling in epidermal stem cells in vitro. TNF-induced β-catenin accumulation is dependent on AKT but not Wnt signalling. Inhibition of PI3K/AKT blocks depilation-induced HF TAT. Notably, Pten loss in Lgr5+ HF stem cells results in HF TAT independent of injury and promotes HF neogenesis after wounding. Thus, our results suggest that macrophage-TNF-induced AKT/β-catenin signalling in Lgr5+ HF stem cells has a crucial role in promoting HF cycling and neogenesis after wounding.
Collapse
Affiliation(s)
- Xusheng Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Haiyan Chen
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China
- State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, 518055 Shenzhen, China
| | - Ruiyun Tian
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
- Shenzhen Peiyuan Biotechnology Company, Shenzhen 518055, China
| | - Yiling Zhang
- Department of Orthopedics, The General Hospital of Chinese People's Liberation Army, Beijing 100039, China
| | - Marina S. Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Chengmei Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Jianfeng Ge
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Zhimeng Fan
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Deqiang Kong
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Xiaoxiao Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Ting Cai
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Ying Zhou
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Jingwen Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Jinmei Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Shan Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Zhihai Qin
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Huanhuan Jia
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou 510260, China
| | - Yue Wu
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou 510260, China
| | - Jia Liu
- College of Life Sciences, Northeast Forestry University, Harbin 100040, China
| | - Sergei A. Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Edward E. Tredget
- Wound Healing Research Group, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada ABT6G2E1
| | - Mei Lin
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Jianjun Liu
- Medical Key Laboratory of Health Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen 518054, China
| | - Yuyang Jiang
- State Key Laboratory Breeding Base-Shenzhen Key Laboratory of Chemical Biology, Graduate School at Shenzhen, Tsinghua University, 518055 Shenzhen, China
| | - Yaojiong Wu
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China
| |
Collapse
|
226
|
Brown JM, Recht L, Strober S. The Promise of Targeting Macrophages in Cancer Therapy. Clin Cancer Res 2017; 23:3241-3250. [PMID: 28341752 DOI: 10.1158/1078-0432.ccr-16-3122] [Citation(s) in RCA: 255] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/17/2017] [Accepted: 03/17/2017] [Indexed: 12/14/2022]
Abstract
Cancer therapy has developed around the concept of killing, or stopping the growth of, the cancer cells. Molecularly targeted therapy is the modern expression of this paradigm. Increasingly, however, the realization that the cancer has co-opted the normal cells of the stroma for its own survival has led to the concept that the tumor microenvironment (TME) could be targeted for effective therapy. In this review, we outline the importance of tumor-associated macrophages (TAM), a major component of the TME, in the response of tumors to cancer therapy. We discuss the normal role of macrophages in wound healing, the major phenotypes of TAMs, and their role in blunting the efficacy of cancer treatment by radiation and anticancer drugs, both by promoting tumor angiogenesis and by suppressing antitumor immunity. Finally, we review the many preclinical studies that have shown that the response of tumors to irradiation and anticancer drugs can be improved, sometimes markedly so, by depleting TAMs from tumors or by suppressing their polarization from an M1 to an M2 phenotype. The data clearly support the validity of clinical testing of combining targeting TAMs with conventional therapy. Clin Cancer Res; 23(13); 3241-50. ©2017 AACR.
Collapse
Affiliation(s)
- J Martin Brown
- Department of Radiation Oncology, Stanford University, Stanford, California.
| | - Lawrence Recht
- Department of Neurology, Stanford University, Stanford, California
| | - Samuel Strober
- Department of Medicine, Stanford University, Stanford, California
| |
Collapse
|
227
|
Ito T, Ishigami M, Matsushita Y, Hirata M, Matsubara K, Ishikawa T, Hibi H, Ueda M, Hirooka Y, Goto H, Yamamoto A. Secreted Ectodomain of SIGLEC-9 and MCP-1 Synergistically Improve Acute Liver Failure in Rats by Altering Macrophage Polarity. Sci Rep 2017; 7:44043. [PMID: 28272428 PMCID: PMC5358744 DOI: 10.1038/srep44043] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/01/2017] [Indexed: 12/21/2022] Open
Abstract
Effective treatments for acute liver failure (ALF) are still lacking. We recently reported that a single intravenous administration of serum-free conditioned medium from stem cells derived from human exfoliated deciduous teeth (SHED-CM) into the D-galactosamine (D-Gal)-induced rat ALF model improves the liver injury. However, the specific factors in SHED-CM that are responsible for resolving ALF remain unclear. Here we found that depleting SHED-CM of two anti-inflammatory M2 macrophage inducers—monocyte chemoattractant protein-1 (MCP-1) and the secreted ectodomain of sialic acid-binding Ig-like lectin-9 (sSiglec-9)—abolished its ability to resolve rat ALF. Furthermore, treatment with MCP-1/sSiglec-9 alone dramatically improved the survival of ALF rats. This treatment induced anti-inflammatory M2, suppressed hepatocyte apoptosis, and promoted hepatocyte proliferation. Treatment with an M2-depletion reagent (mannosylated clodronate liposomes) suppressed the recovery. In addition, MCP-1 and sSiglec-9 synergistically promoted the M2 differentiation of bone marrow-derived macrophages via CCR2, accompanied by the production of multiple liver-regenerating factors. The conditioned medium from MCP-1/sSiglec-9-activated M2 macrophages, but not from interleukin-4-induced ones, suppressed the D-Gal- and LPS-induced apoptosis of primary hepatocytes and promoted their proliferation in vitro. The unique combination of MCP-1/sSiglec-9 ameliorates rat ALF by inhibiting hepatocellular apoptosis and promoting liver regeneration through the induction of anti-inflammatory/tissue-repairing M2 macrophages.
Collapse
Affiliation(s)
- Takanori Ito
- Department of Gastroenterology and Hepatology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Masatoshi Ishigami
- Department of Gastroenterology and Hepatology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yoshihiro Matsushita
- Department of Oral and Maxillofacial Surgery of Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Marina Hirata
- Department of Oral and Maxillofacial Surgery of Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Kohki Matsubara
- Department of Oral and Maxillofacial Surgery of Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Tetsuya Ishikawa
- Department of Gastroenterology and Hepatology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hideharu Hibi
- Department of Oral and Maxillofacial Surgery of Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Minoru Ueda
- Department of Oral and Maxillofacial Surgery of Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yoshiki Hirooka
- Department of Gastroenterology and Hepatology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hidemi Goto
- Department of Gastroenterology and Hepatology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Akihito Yamamoto
- Department of Oral and Maxillofacial Surgery of Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.,Department of Oral histology, Institute of Biomedical Science, Tokushima University Graduate School, 3-18-5 Kuramoto-cho, Tokushima 770-8504, Japan
| |
Collapse
|
228
|
Hosur V, Burzenski LM, Stearns TM, Farley ML, Sundberg JP, Wiles MV, Shultz LD. Early induction of NRF2 antioxidant pathway by RHBDF2 mediates rapid cutaneous wound healing. Exp Mol Pathol 2017; 102:337-346. [PMID: 28268192 DOI: 10.1016/j.yexmp.2017.03.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 03/01/2017] [Indexed: 12/16/2022]
Abstract
Rhomboid family protein RHBDF2, an upstream regulator of the epidermal growth factor (EGF) receptor signaling, has been implicated in cutaneous wound healing. However, the underlying molecular mechanisms are still emerging. In humans, a gain-of-function mutation in the RHBDF2 gene accelerates cutaneous wound healing in an EGFR-dependent manner. Likewise, a gain-of-function mutation in the mouse Rhbdf2 gene (Rhbdf2cub/cub) shows a regenerative phenotype (rapid ear-hole closure) resulting from constitutive activation of the EGFR pathway. Because the RHBDF2-regulated EGFR pathway is relevant to cutaneous wound healing in humans, we used Rhbdf2cub/cub mice to investigate the biological networks and pathways leading to accelerated ear-hole closure, with the goal of identifying therapeutic targets potentially effective in promoting wound healing in humans. Comparative transcriptome analysis of ear pinna tissue from Rhbdf2cub/cub and Rhbdf2+/+ mice at 0h, 15min, 2h, and 24h post-wounding revealed an early induction of the nuclear factor E2-related factor 2 (NRF2)-mediated anti-oxidative pathway (0h and 15min), followed by the integrin-receptor aggregation pathway (2h) as early-stage events immediately and shortly after wounding in Rhbdf2cub/cub mice. Additionally, we observed genes enriched for the Fc fragment of the IgG receptor IIIa (FCGR3A)-mediated phagocytosis pathway 24h post-wounding. Although cutaneous wound repair in healthy individuals is generally non-problematic, it can be severely impaired due to aging, diabetes, and chronic inflammation. This study suggests that activation of the NRF2-antioxidant pathway by rhomboid protein RHBDF2 might be beneficial in treating chronic non-healing wounds.
Collapse
Affiliation(s)
- Vishnu Hosur
- The Jackson Laboratory, Bar Harbor, ME 04609, United States.
| | | | | | | | | | | | | |
Collapse
|
229
|
Herter EK, Xu Landén N. Non-Coding RNAs: New Players in Skin Wound Healing. Adv Wound Care (New Rochelle) 2017; 6:93-107. [PMID: 28289554 PMCID: PMC5346954 DOI: 10.1089/wound.2016.0711] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 11/26/2016] [Indexed: 12/22/2022] Open
Abstract
Significance: Wound healing is a basic physiological process that is utilized to keep the integrity of the skin. Impaired wound repair, such as chronic wounds and pathological scars, presents a major health and economic burden worldwide. To date, efficient targeted treatment for these wound disorders is still lacking, which is largely due to our limited understanding of the biological mechanisms underlying these diseases. Research driven around discovering new therapies for these complications is, therefore, an urgent need. Recent Advances: The vast majority of the human genome is transcribed to RNAs that lack protein-coding capacity. Intensive research in the recent decade has revealed that these non-coding RNAs (ncRNAs) function as important regulators of cellular physiology and pathology, which makes them promising therapeutic and diagnostic entities. Critical Issues: A class of short ncRNAs, microRNAs, has been found to be indispensable for all the phases of skin wound healing and plays important roles in the pathogenesis of wound complications. The role of long ncRNAs (lncRNA) in skin wound healing remains largely unexplored. Recent studies revealed the essential role of lncRNAs in epidermal differentiation and stress response, indicating their potential importance for skin wound healing, which warrants future research. Future Directions: An investigation of ncRNAs will add new layers of complexity to our understanding of normal skin wound healing as well as to the pathogenesis of wound disorders. Development of ncRNA-based biomarkers and treatments is an interesting and important avenue for future research on wound healing.
Collapse
Affiliation(s)
- Eva K. Herter
- Unit of Dermatology and Venereology, Molecular Dermatology Research Group, Department of Medicine, Center for Molecular Medicine (CMM), Karolinska Institutet, Stockholm, Sweden
| | - Ning Xu Landén
- Unit of Dermatology and Venereology, Molecular Dermatology Research Group, Department of Medicine, Center for Molecular Medicine (CMM), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
230
|
Makrantonaki E, Wlaschek M, Scharffetter-Kochanek K. Pathogenese von Wundheilungsstörungen bei älteren Patienten. J Dtsch Dermatol Ges 2017; 15:255-278. [DOI: 10.1111/ddg.13199_g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/21/2016] [Indexed: 01/13/2023]
|
231
|
Baticic Pucar L, Pernjak Pugel E, Detel D, Varljen J. Involvement of DPP IV/CD26 in cutaneous wound healing process in mice. Wound Repair Regen 2017; 25:25-40. [PMID: 27868279 DOI: 10.1111/wrr.12498] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 11/14/2016] [Indexed: 12/26/2022]
Abstract
Dipeptidyl peptidase IV (DPP IV/CD26) is a widely distributed multifunctional protein that plays a significant role in different physiological as well as pathological processes having a broad spectrum of bioactive substrates and immunomodulative properties. It has potential influence on different processes crucial for wound healing, including cell adhesion, migration, apoptosis, and extracellular matrix degradation. However, despite its known enzymatic and immunomodulative functions, limited data characterize the role of DPP IV/CD26 in cutaneous wound healing mechanisms. The aim of this study was to investigate the process of wound healing in conditions of CD26 deficiency in order to obtain better insights on the role of DPP IV/CD26 in cutaneous regeneration. Experimental wounds were made on the dorsal part of CD26 deficient (CD26-/- ) and wild-type mice (C57BL/6). The process of cutaneous wound healing was monitored on defined time schedule postwounding by macroscopic, microscopic, and biochemical analyses. Obtained results revealed a better rate of wound closure, revascularization and cell proliferation in CD26-/- mice, with enhanced local expression of hypoxia-inducible factor 1α and vascular endothelial growth factor. CD26 deficiency induced prompt macrophage recruitment at the site of skin damage but did not influence mobilization of T-cells in comparison with wild-type mice. CD26-/- mice have significantly higher values of IP-10 in serum and control skins compared with wild-type mice but values in wounds did not differ significantly on days 2, 4, and 7 of wound healing. DPP IV/CD26 activity was found to be decreased 4 days postwounding in serum and 2, 4, and 7 days postwounding in wounds of wild-type animals compared with control skins. These findings contribute to better understanding of wound healing mechanisms and could give a support in finding new therapeutic approaches for wound healing and tissue regeneration.
Collapse
Affiliation(s)
- Lara Baticic Pucar
- Department of Chemistry and Biochemistry, Faculty of Medicine, University of Rijeka, Brace Branchetta 20, Rijeka, 51000, Hrvatska-Croatia
| | - Ester Pernjak Pugel
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Hrvatska-Croatia
| | - Dijana Detel
- Department of Chemistry and Biochemistry, Faculty of Medicine, University of Rijeka, Brace Branchetta 20, Rijeka, 51000, Hrvatska-Croatia
| | - Jadranka Varljen
- Department of Chemistry and Biochemistry, Faculty of Medicine, University of Rijeka, Brace Branchetta 20, Rijeka, 51000, Hrvatska-Croatia
| |
Collapse
|
232
|
Ridiandries A, Bursill C, Tan J. Broad-Spectrum Inhibition of the CC-Chemokine Class Improves Wound Healing and Wound Angiogenesis. Int J Mol Sci 2017; 18:ijms18010155. [PMID: 28098795 PMCID: PMC5297788 DOI: 10.3390/ijms18010155] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 01/08/2023] Open
Abstract
Angiogenesis is involved in the inflammation and proliferation stages of wound healing, to bring inflammatory cells to the wound and provide a microvascular network to maintain new tissue formation. An excess of inflammation, however, leads to prolonged wound healing and scar formation, often resulting in unfavourable outcomes such as amputation. CC-chemokines play key roles in the promotion of inflammation and inflammatory-driven angiogenesis. Therefore, inhibition of the CC-chemokine class may improve wound healing. We aimed to determine if the broad-spectrum CC-chemokine inhibitor “35K” could accelerate wound healing in vivo in mice. In a murine wound healing model, 35K protein or phosphate buffered saline (PBS, control) were added topically daily to wounds. Cohorts of mice were assessed in the early stages (four days post-wounding) and in the later stages of wound repair (10 and 21 days post-wounding). Topical application of the 35K protein inhibited CC-chemokine expression (CCL5, CCL2) in wounds and caused enhanced blood flow recovery and wound closure in early-mid stage wounds. In addition, 35K promoted neovascularisation in the early stages of wound repair. Furthermore, 35K treated wounds had significantly lower expression of the p65 subunit of NF-κB, a key inflammatory transcription factor, and augmented wound expression of the pro-angiogenic and pro-repair cytokine TGF-β. These findings show that broad-spectrum CC-chemokine inhibition may be beneficial for the promotion of wound healing.
Collapse
Affiliation(s)
- Anisyah Ridiandries
- Heart Research Institute, 7 Eliza Street, Newtown, Sydney 2042, NSW, Australia.
- Sydney Medical School, University of Sydney, Camperdown, Sydney 2050, NSW, Australia.
| | - Christina Bursill
- Heart Research Institute, 7 Eliza Street, Newtown, Sydney 2042, NSW, Australia.
- Sydney Medical School, University of Sydney, Camperdown, Sydney 2050, NSW, Australia.
| | - Joanne Tan
- Heart Research Institute, 7 Eliza Street, Newtown, Sydney 2042, NSW, Australia.
- Sydney Medical School, University of Sydney, Camperdown, Sydney 2050, NSW, Australia.
| |
Collapse
|
233
|
Hsieh JY, Smith TD, Meli VS, Tran TN, Botvinick EL, Liu WF. Differential regulation of macrophage inflammatory activation by fibrin and fibrinogen. Acta Biomater 2017; 47:14-24. [PMID: 27662809 DOI: 10.1016/j.actbio.2016.09.024] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/12/2016] [Accepted: 09/19/2016] [Indexed: 12/20/2022]
Abstract
Fibrin is a major component of the provisional extracellular matrix formed during tissue repair following injury, and enables cell infiltration and anchoring at the wound site. Macrophages are dynamic regulators of this process, advancing and resolving inflammation in response to cues in their microenvironment. Although much is known about how soluble factors such as cytokines and chemokines regulate macrophage polarization, less is understood about how insoluble and adhesive cues, specifically the blood coagulation matrix fibrin, influence macrophage behavior. In this study, we observed that fibrin and its precursor fibrinogen elicit distinct macrophage functions. Culturing macrophages on fibrin gels fabricated by combining fibrinogen with thrombin stimulated secretion of the anti-inflammatory cytokine, interleukin-10 (IL-10). In contrast, exposure of macrophages to soluble fibrinogen stimulated high levels of inflammatory cytokine tumor necrosis factor alpha (TNF-α). Macrophages maintained their anti-inflammatory behavior when cultured on fibrin gels in the presence of soluble fibrinogen. In addition, adhesion to fibrin matrices inhibited TNF-α production in response to stimulation with LPS and IFN-γ, cytokines known to promote inflammatory macrophage polarization. Our data demonstrate that fibrin exerts a protective effect on macrophages, preventing inflammatory activation by stimuli including fibrinogen, LPS, and IFN-γ. Together, our study suggests that the presentation of fibrin(ogen) may be a key switch in regulating macrophage phenotype behavior, and this feature may provide a valuable immunomodulatory strategy for tissue healing and regeneration. STATEMENT OF SIGNIFICANCE Fibrin is a fibrous protein resulting from blood clotting and provides a provisional matrix into which cells migrate and to which they adhere during wound healing. Macrophages play an important role in this process, and are needed for both advancing and resolving inflammation. We demonstrate that culture of macrophages on fibrin matrices exerts an anti-inflammatory effect, whereas the soluble precursor fibrinogen stimulates inflammatory activation. Moreover, culture on fibrin completely abrogates inflammatory signaling caused by fibrinogen or known inflammatory stimuli including LPS and IFN-γ. Together, these studies show that the presentation of fibrin(ogen) is important for regulating a switch between macrophage pro- and anti-inflammatory behavior.
Collapse
Affiliation(s)
- Jessica Y Hsieh
- Department of Biomedical Engineering, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States; Department of Chemical Engineering and Materials Science, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States
| | - Tim D Smith
- Department of Biomedical Engineering, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States; Department of Chemical Engineering and Materials Science, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States
| | - Vijaykumar S Meli
- Department of Biomedical Engineering, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States; Department of Chemical Engineering and Materials Science, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States
| | - Thi N Tran
- Department of Biomedical Engineering, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States; Department of Chemical Engineering and Materials Science, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States
| | - Elliot L Botvinick
- Department of Biomedical Engineering, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States; Department of Chemical Engineering and Materials Science, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States
| | - Wendy F Liu
- Department of Biomedical Engineering, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States; Department of Chemical Engineering and Materials Science, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, 2412 Engineering Hall, Irvine, CA 92697-2730, United States.
| |
Collapse
|
234
|
Abstract
Macrophages are the initial biologic responders to biomaterials. These highly plastic immune sentinels control and modulate responses to materials, foreign or natural. The responses may vary from immune stimulatory to immune suppressive. Several parameters have been identified that influence macrophage response to biomaterials, specifically size, geometry, surface topography, hydrophobicity, surface chemistry, material mechanics, and protein adsorption. In this review, the influence of these parameters is supported with examples of both synthetic and naturally derived materials and illustrates that a combination of these parameters ultimately influences macrophage responses to the biomaterial. Having an understanding of these properties may lead to highly efficient design of biomaterials with desirable biologic response properties.
Collapse
|
235
|
Maruyama Y, Inoue K, Mori K, Gorai K, Shimamoto R, Onitsuka T, Iguchi H, Okazaki M, Nakagawa M. Neutrophil-lymphocyte ratio and platelet-lymphocyte ratio as predictors of wound healing failure in head and neck reconstruction. Acta Otolaryngol 2017; 137:106-110. [PMID: 27553628 DOI: 10.1080/00016489.2016.1218047] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CONCLUSIONS In microsurgical head and neck reconstruction, a higher rate of post-operative wound complication could be predicted by a lower pre-operative neutrophil ratio (< 64.9%), neutrophil-lymphocyte ratio (NLR) (< 3.5), and platelet-lymphocyte ratio (PLR) (< 160). OBJECTIVES To evaluate the predictor of post-operative wound complications in microsurgical head and neck reconstruction. METHODS Patients who were undergoing tumor ablation and microsurgical reconstruction from April 2011 to March 2014 were analyzed retrospectively. The pre-operative hematological data, age, sex, co-morbidities, body mass index (BMI), adjuvant therapies, smoking, operation time, blood loss, total protein, T-stage, and Anesthesiologists Performance Status (ASA-PS) score were collected. Cases of post-operative wound healing failure were reviewed. RESULTS One hundred and three consecutive patients were enrolled. Among these, the results of 77 patients who were younger than 70 years of age were analyzed. The distributions of the neutrophil ratio (p = .0005), lymphocyte ratio (p = .0166), monocyte ratio (p = .0341), NLR (p = .005), and PLR (p = .008) differed significantly between the patients with and without post-operative wound healing failure. Neutrophil ratio, NLR, and PLR cut-off values of 64.9, 3.5, and 160 were significantly associated with the rate of wound healing failure rate (p = .0002, .00021, .0042, respectively).
Collapse
|
236
|
Wang Q, Zhu G, Cao X, Dong J, Song F, Niu Y. Blocking AGE-RAGE Signaling Improved Functional Disorders of Macrophages in Diabetic Wound. J Diabetes Res 2017; 2017:1428537. [PMID: 29119117 PMCID: PMC5651124 DOI: 10.1155/2017/1428537] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 08/17/2017] [Indexed: 11/21/2022] Open
Abstract
Advanced glycosylation end products (AGEs) accumulate in diabetic wounds. Interactions between AGEs and their receptor (RAGE) leads to dermatologic problems in diabetes. Macrophage, which plays important roles in wound healing, highly expresses RAGE. Therefore, we investigated whether RAGE-expressing macrophages might be responsible for impaired wound healing on diabetes. We used anti-RAGE antibody applied topically on diabetic wounds. After confirming that wound healing was improved in anti-RAGE antibody group compared with normal mice, our results showed that macrophages appeared insufficient in the early stage and fading away slowly in the later proliferative phase compared with the control group, which was ameliorated in anti-RAGE antibody-applied wounds. Blocking AGE-RAGE signaling also increased neutrophils phagocytized by macrophages and promoted the phenotypic switch of macrophages from proinflammatory to prohealing activities. In vitro, phagocytosis of THP-1 (M0) and lipopolysaccharide- (LPS-) induced (M1) macrophages was impaired by treatment with AGEs, while IL-4- and IL-13-induced (M2) macrophages was not. Finally, AGEs increased the proinflammatory response of M1 macrophages, while inhibiting the polarization and anti-inflammatory functions of M2 macrophages. In conclusion, inhibition of AGE-RAGE signaling improved functional disorders of macrophages in the early inflammatory phase, which promoted the healing of wounds in diabetic mice.
Collapse
Affiliation(s)
- Qi Wang
- Shanghai Burn Institute, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanya Zhu
- Shanghai Burn Institute, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaozan Cao
- Shanghai Burn Institute, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaoyun Dong
- Shanghai Burn Institute, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Song
- Shanghai Burn Institute, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwen Niu
- Shanghai Burn Institute, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
237
|
Abstract
Chronic wounds pose considerable public health challenges and burden. Wound healing is known to require the participation of macrophages, but mechanisms remain unclear. The M1 phenotype macrophages have a known scavenger function, but they also play multiple roles in tissue repair and regeneration when they transition to an M2 phenotype. Macrophage precursors (mononuclear cells/monocytes) follow the influx of PMN neutrophils into a wound during the natural wound-healing process, to become the major cells in the wound. Natural wound-healing process is a four-phase progression consisting of hemostasis, inflammation, proliferation, and remodeling. A lag phase of 3-6 days precedes the remodeling phase, which is characterized by fibroblast activation and finally collagen production. This normal wound-healing process can be accelerated by the intracellular delivery of ATP to wound tissue. This novel ATP-mediated acceleration arises due to an alternative activation of the M1 to M2 transition (macrophage polarization), a central and critical feature of the wound-healing process. This response is also characterized by an early increased release of pro-inflammatory cytokines (TNF, IL-1 beta, IL-6), a chemokine (MCP-1), an activation of purinergic receptors (a family of plasma membrane receptors found in almost all mammalian cells), and an increased production of platelets and platelet microparticles. These factors trigger a massive influx of macrophages, as well as in situ proliferation of the resident macrophages and increased synthesis of VEGFs. These responses are followed, in turn, by rapid neovascularization and collagen production by the macrophages, resulting in wound covering with granulation tissue within 24 h.
Collapse
Affiliation(s)
| | - Sufan Chien
- Noveratech LLC, Louisville, KY, USA.
- Department of Surgery, University of Louisville, School of Medicine, Louisville, KY, USA.
| |
Collapse
|
238
|
Sorg H, Tilkorn DJ, Hager S, Hauser J, Mirastschijski U. Skin Wound Healing: An Update on the Current Knowledge and Concepts. Eur Surg Res 2016; 58:81-94. [PMID: 27974711 DOI: 10.1159/000454919] [Citation(s) in RCA: 707] [Impact Index Per Article: 78.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 12/05/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND The integrity of healthy skin plays a crucial role in maintaining physiological homeostasis of the human body. The skin is the largest organ system of the body. As such, it plays pivotal roles in the protection against mechanical forces and infections, fluid imbalance, and thermal dysregulation. At the same time, it allows for flexibility to enable joint function in some areas of the body and more rigid fixation to hinder shifting of the palm or foot sole. Many instances lead to inadequate wound healing which necessitates medical intervention. Chronic conditions such as diabetes mellitus or peripheral vascular disease can lead to impaired wound healing. Acute trauma such as degloving or large-scale thermal injuries are followed by a loss of skin organ function rendering the organism vulnerable to infections, thermal dysregulation, and fluid loss. METHODS For this update article, we have reviewed the actual literature on skin wound healing purposes focusing on the main phases of wound healing, i.e., inflammation, proliferation, epithelialization, angiogenesis, remodeling, and scarring. RESULTS The reader will get briefed on new insights and up-to-date concepts in skin wound healing. The macrophage as a key player in the inflammatory phase will be highlighted. During the epithelialization process, we will present the different concepts of how the wound will get closed, e.g., leapfrogging, lamellipodial crawling, shuffling, and the stem cell niche. The neovascularization represents an essential component in wound healing due to its fundamental impact from the very beginning after skin injury until the end of the wound remodeling. Here, the distinct pattern of the neovascularization process and the special new functions of the pericyte will be underscored. At the end, this update will present 3 topics of high interest in skin wound healing issues, dealing with scarring, tissue engineering, and plasma application. CONCLUSION Although wound healing mechanisms and specific cell functions in wound repair have been delineated in part, many underlying pathophysiological processes are still unknown. The purpose of the following update on skin wound healing is to focus on the different phases and to brief the reader on the current knowledge and new insights. Skin wound healing is a complex process, which is dependent on many cell types and mediators interacting in a highly sophisticated temporal sequence. Although some interactions during the healing process are crucial, redundancy is high and other cells or mediators can adopt functions or signaling without major complications.
Collapse
Affiliation(s)
- Heiko Sorg
- Department of Plastic, Reconstructive and Aesthetic Surgery, Hand Surgery, Alfried Krupp Krankenhaus Essen, Essen, Germany
| | | | | | | | | |
Collapse
|
239
|
Kano F, Matsubara K, Ueda M, Hibi H, Yamamoto A. Secreted Ectodomain of Sialic Acid-Binding Ig-Like Lectin-9 and Monocyte Chemoattractant Protein-1 Synergistically Regenerate Transected Rat Peripheral Nerves by Altering Macrophage Polarity. Stem Cells 2016; 35:641-653. [PMID: 27862629 DOI: 10.1002/stem.2534] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/30/2016] [Accepted: 10/10/2016] [Indexed: 12/24/2022]
Abstract
Peripheral nerves (PNs) exhibit remarkable self-repairing reparative activity after a simple crush or cut injury. However, the neuronal transection involving a nerve gap overwhelms their repairing activity and causes persistent paralysis. Here, we show that an implantation of the serum-free conditioned medium from stem cells from human exfoliated deciduous teeth (SHED-CM) immersed in a collagen sponge into the nerve gap formed by rat facial nerves transection restored the neurological function. In contrast, SHED-CM specifically depleted of a set of anti-inflammatory M2 macrophage inducers, monocyte chemoattractant protein-1 (MCP-1) and the secreted ectodomain of sialic acid-binding Ig-like lectin-9 (sSiglec-9) lost the ability to restore neurological function in this model. Notably, the combination of MCP-1 and sSiglec-9 induced the polarization of M2 macrophages in vitro, resulting in the expression of multiple trophic factors that enhanced proliferation, migration, and differentiation of Schwann cells, blood vessel formation, and nerve fiber extension. Furthermore, the implantation of a collagen graft containing MCP-1/sSiglec-9 into the nerve gap induced anti-inflammatory M2 macrophage polarization, generated a Schwann-cell bridge instead of fibrotic scar, induced axonal regrowth, and restored nerve function. The specific elimination of M2 macrophages by Mannosylated-Clodrosome suppressed the MCP-1/sSiglec-9-mediated neurological recovery. Taken together, our data suggest that MCP-1/sSiglec-9 regenerates PNs by inducing tissue-repairing M2 macrophages and may provide therapeutic benefits for severe peripheral nerve injuries. Stem Cells 2017;35:641-653.
Collapse
Affiliation(s)
- Fumiya Kano
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Kohki Matsubara
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Minoru Ueda
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Hideharu Hibi
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Akihito Yamamoto
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| |
Collapse
|
240
|
Huang YC, Lew WZ, Feng SW, Lai WY, Abiko Y, Huang HM. Histomorphometric and transcriptome evaluation of early healing bone treated with a novel human particulate dentin powder. Biomed Mater 2016; 12:015004. [DOI: 10.1088/1748-605x/12/1/015004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
241
|
Rucavado A, Nicolau CA, Escalante T, Kim J, Herrera C, Gutiérrez JM, Fox JW. Viperid Envenomation Wound Exudate Contributes to Increased Vascular Permeability via a DAMPs/TLR-4 Mediated Pathway. Toxins (Basel) 2016; 8:toxins8120349. [PMID: 27886127 PMCID: PMC5198544 DOI: 10.3390/toxins8120349] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/15/2016] [Accepted: 11/17/2016] [Indexed: 01/21/2023] Open
Abstract
Viperid snakebite envenomation is characterized by inflammatory events including increase in vascular permeability. A copious exudate is generated in tissue injected with venom, whose proteomics analysis has provided insights into the mechanisms of venom-induced tissue damage. Hereby it is reported that wound exudate itself has the ability to induce increase in vascular permeability in the skin of mice. Proteomics analysis of exudate revealed the presence of cytokines and chemokines, together with abundant damage associated molecular pattern molecules (DAMPs) resulting from both proteolysis of extracellular matrix and cellular lysis. Moreover, significant differences in the amounts of cytokines/chemokines and DAMPs were detected between exudates collected 1 h and 24 h after envenomation, thus highlighting a complex temporal dynamic in the composition of exudate. Pretreatment of mice with Eritoran, an antagonist of Toll-like receptor 4 (TLR4), significantly reduced the exudate-induced increase in vascular permeability, thus suggesting that DAMPs might be acting through this receptor. It is hypothesized that an "Envenomation-induced DAMPs cycle of tissue damage" may be operating in viperid snakebite envenomation through which venom-induced tissue damage generates a variety of DAMPs which may further expand tissue alterations.
Collapse
Affiliation(s)
- Alexandra Rucavado
- Instituto Clodomiro Picado, Facultad de Microbiología Universidad de Costa Rica, San José 11501-2060, Costa Rica.
| | - Carolina A Nicolau
- Laboratório de Toxinologia, Instituto Oswaldo Cruz, Rio de Janeiro CEP 21040-360, Brazil.
| | - Teresa Escalante
- Instituto Clodomiro Picado, Facultad de Microbiología Universidad de Costa Rica, San José 11501-2060, Costa Rica.
| | - Junho Kim
- Department of Fine Chemistry & New Materials, Sangji University, Wonju-si, Kangwon-do 220-702, Korea.
| | - Cristina Herrera
- Instituto Clodomiro Picado, Facultad de Microbiología Universidad de Costa Rica, San José 11501-2060, Costa Rica.
- Facultad de Farmacia, Universidad de Costa Rica, San José 11501-2060, Costa Rica.
| | - José María Gutiérrez
- Instituto Clodomiro Picado, Facultad de Microbiología Universidad de Costa Rica, San José 11501-2060, Costa Rica.
| | - Jay W Fox
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, P.O. Box 800734, Charlottesville, VA 22908, USA.
| |
Collapse
|
242
|
Hinz B. The role of myofibroblasts in wound healing. Curr Res Transl Med 2016; 64:171-177. [PMID: 27939455 DOI: 10.1016/j.retram.2016.09.003] [Citation(s) in RCA: 206] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 09/22/2016] [Indexed: 12/20/2022]
Abstract
The importance of proper skin wound healing becomes evident when our body's repair mechanisms fail, leading to either non-healing (chronic) wounds or excessive repair (fibrosis). Chronic wounds are a tremendous burden for patients and global healthcare systems and are on the rise due to their increasing incidence with age and diabetes. Curiously, these same risk factors also sign responsible for the development of hypertrophic scarring and organ fibrosis. Activated repair cells - myofibroblasts - are the main producers and organizers of extracellular matrix which is needed to restore tissue integrity after injury. Too many myofibroblasts working for too long cause tissue contractures that ultimately obstruct organ function. Insufficient myofibroblast activation and activities, in turn, prevents normal wound healing. This short review puts a spotlight on the myofibroblast for those who seek therapeutic targets in the context of dysregulated tissue repair. "Keep your myofibroblasts in balance" is the message.
Collapse
Affiliation(s)
- B Hinz
- Laboratory of tissue repair and regeneration, Matrix dynamics group, faculty of dentistry, university of Toronto, 150, College Street, FitzGerald building, room 234, M5S 3E2 Toronto, Ontario, Canada.
| |
Collapse
|
243
|
Kratofil RM, Kubes P, Deniset JF. Monocyte Conversion During Inflammation and Injury. Arterioscler Thromb Vasc Biol 2016; 37:35-42. [PMID: 27765768 DOI: 10.1161/atvbaha.116.308198] [Citation(s) in RCA: 324] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/10/2016] [Indexed: 12/25/2022]
Abstract
Monocytes are circulating leukocytes important in both innate and adaptive immunity, primarily functioning in immune defense, inflammation, and tissue remodeling. There are 2 subsets of monocytes in mice (3 subsets in humans) that are mobilized from the bone marrow and recruited to sites of inflammation, where they carry out their respective functions in promoting inflammation or facilitating tissue repair. Our understanding of the fate of these monocyte subsets at the site of inflammation is constantly evolving. This brief review highlights the plasticity of monocyte subsets and their conversion during inflammation and injury.
Collapse
Affiliation(s)
- Rachel M Kratofil
- From the Department of Microbiology, Immunology, and Infectious Diseases (R.M.K., P.K.) and Department of Physiology and Pharmacology (P.K., J.F.D.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Canada
| | - Paul Kubes
- From the Department of Microbiology, Immunology, and Infectious Diseases (R.M.K., P.K.) and Department of Physiology and Pharmacology (P.K., J.F.D.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Canada
| | - Justin F Deniset
- From the Department of Microbiology, Immunology, and Infectious Diseases (R.M.K., P.K.) and Department of Physiology and Pharmacology (P.K., J.F.D.), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Canada
| |
Collapse
|
244
|
Abstract
Macrophages are heterogeneous cells that play a key role in inflammatory and tissue reparative responses. Over the past decade it has become clear that shifts in cellular metabolism are important determinants of macrophage function and phenotype. At the same time, our appreciation of macrophage diversity in vivo has also been increasing. Factors such as cell origin and tissue localization are now recognized as important variables that influence macrophage biology. Whether different macrophage populations also have unique metabolic phenotypes has not been extensively explored. In this article, we will discuss the importance of understanding how macrophage origin can modulate metabolic programming and influence inflammatory responses.
Collapse
|
245
|
Hosono K, Isonaka R, Kawakami T, Narumiya S, Majima M. Signaling of Prostaglandin E Receptors, EP3 and EP4 Facilitates Wound Healing and Lymphangiogenesis with Enhanced Recruitment of M2 Macrophages in Mice. PLoS One 2016; 11:e0162532. [PMID: 27711210 PMCID: PMC5053515 DOI: 10.1371/journal.pone.0162532] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 08/24/2016] [Indexed: 01/09/2023] Open
Abstract
Lymphangiogenesis plays an important role in homeostasis, metabolism, and immunity, and also occurs during wound-healing. Here, we examined the roles of prostaglandin E2 (PGE2) receptor (EP) signaling in enhancement of lymphangiogenesis in wound healing processes. The hole-punch was made in the ears of male C57BL/6 mice using a metal ear punch. Healing process and lymphangiogenesis together with macrophage recruitment were analyzed in EP knockout mice. Lymphangiogenesis was up-regulated in the granulation tissues at the margins of punched-hole wounds in mouse ears, and this increase was accompanied by increased expression levels of COX-2 and microsomal prostaglandin E synthase-1. Administration of celecoxib, a COX-2 inhibitor, suppressed lymphangiogenesis in the granulation tissues and reduced the induction of the pro-lymphangiogenic factors, vascular endothelial growth factor (VEGF) -C and VEGF-D. Topical applications of selective EP receptor agonists enhanced the expressions of lymphatic vessel endothelial hyaluronan receptor-1 and VEGF receptor-3. The wound-healing processes and recruitment of CD11b-positive macrophages, which produced VEGF-C and VEGF-D, were suppressed under COX-2 inhibition. Mice lacking either EP3 or EP4 exhibited reduced wound-healing, lymphangiogenesis and recruitment of M2 macrophages, compared with wild type mice. Proliferation of cultured human lymphatic endothelial cells was not detected under PGE2 stimulation. Lymphangiogenesis and recruitment of M2 macrophages that produced VEGF-C/D were suppressed in mice treated with a COX-2 inhibitor or lacking either EP3 or EP4 during wound healing. COX-2 and EP3/EP4 signaling may be novel targets to control lymphangiogenesis in vivo.
Collapse
MESH Headings
- Animals
- CD11b Antigen/metabolism
- Cyclooxygenase 2/metabolism
- Cyclooxygenase 2 Inhibitors/pharmacology
- Ear/physiology
- Gene Knockout Techniques
- Lymphangiogenesis/drug effects
- Macrophages/cytology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Prostaglandin-E Synthases/metabolism
- Receptors, Prostaglandin E, EP3 Subtype/deficiency
- Receptors, Prostaglandin E, EP3 Subtype/genetics
- Receptors, Prostaglandin E, EP3 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/deficiency
- Receptors, Prostaglandin E, EP4 Subtype/genetics
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Signal Transduction/drug effects
- Up-Regulation/drug effects
- Vascular Endothelial Growth Factor C/biosynthesis
- Vascular Endothelial Growth Factor D/biosynthesis
- Wound Healing/drug effects
Collapse
Affiliation(s)
- Kanako Hosono
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Risa Isonaka
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Tadashi Kawakami
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Shuh Narumiya
- Center for Innovation in Immunoregulation Technology and Therapeutics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masataka Majima
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
- * E-mail:
| |
Collapse
|
246
|
Franklin BM, Maroudas E, Osborn JL. Sine-wave electrical stimulation initiates a voltage-gated potassium channel-dependent soft tissue response characterized by induction of hemocyte recruitment and collagen deposition. Physiol Rep 2016; 4:4/12/e12832. [PMID: 27335435 PMCID: PMC4923233 DOI: 10.14814/phy2.12832] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/25/2016] [Indexed: 12/13/2022] Open
Abstract
Soft tissue repair is a complex process that requires specific communication between multiple cell types to orchestrate effective restoration of physiological functions. Macrophages play a critical role in this wound healing process beginning at the onset of tissue injury. Understanding the signaling mechanisms involved in macrophage recruitment to the wound site is an essential step for developing more effective clinical therapies. Macrophages are known to respond to electrical fields, but the underlying cellular mechanisms mediating this response is unknown. This study demonstrated that low‐amplitude sine‐wave electrical stimulation (ES) initiates a soft tissue response in the absence of injury in Procambarus clarkii. This cellular response was characterized by recruitment of macrophage‐like hemocytes to the stimulation site indicated by increased hemocyte density at the site. ES also increased tissue collagen deposition compared to sham treatment (P < 0.05). Voltage‐gated potassium (KV) channel inhibition with either 4‐aminopyridine or astemizole decreased both hemocyte recruitment and collagen deposition compared to saline infusion (P < 0.05), whereas inhibition of calcium‐permeable channels with ruthenium red did not affect either response to ES. Thus, macrophage‐like hemocytes in P. clarkii elicit a wound‐like response to exogenous ES and this is accompanied by collagen deposition. This response is mediated by KV channels but independent of Ca2+ channels. We propose a significant role for KV channels that extends beyond facilitating Ca2+ transport via regulation of cellular membrane potentials during ES of soft tissue.
Collapse
Affiliation(s)
| | - Eleni Maroudas
- Department of Biology, University of Kentucky, Lexington, Kentucky
| | - Jeffrey L Osborn
- Department of Biology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
247
|
Hu K, Olsen BR. The roles of vascular endothelial growth factor in bone repair and regeneration. Bone 2016; 91:30-8. [PMID: 27353702 PMCID: PMC4996701 DOI: 10.1016/j.bone.2016.06.013] [Citation(s) in RCA: 412] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/22/2016] [Accepted: 06/23/2016] [Indexed: 02/08/2023]
Abstract
Vascular endothelial growth factor-A (VEGF) is one of the most important growth factors for regulation of vascular development and angiogenesis. Since bone is a highly vascularized organ and angiogenesis plays an important role in osteogenesis, VEGF also influences skeletal development and postnatal bone repair. Compromised bone repair and regeneration in many patients can be attributed to impaired blood supply; thus, modulation of VEGF levels in bones represents a potential strategy for treating compromised bone repair and improving bone regeneration. This review (i) summarizes the roles of VEGF at different stages of bone repair, including the phases of inflammation, endochondral ossification, intramembranous ossification during callus formation and bone remodeling; (ii) discusses different mechanisms underlying the effects of VEGF on osteoblast function, including paracrine, autocrine and intracrine signaling during bone repair; (iii) summarizes the role of VEGF in the bone regenerative procedure, distraction osteogenesis; and (iv) reviews evidence for the effects of VEGF in the context of repair and regeneration techniques involving the use of scaffolds, skeletal stem cells and growth factors.
Collapse
Affiliation(s)
- Kai Hu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA.
| | - Bjorn R Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA.
| |
Collapse
|
248
|
Klopfleisch R. Macrophage reaction against biomaterials in the mouse model - Phenotypes, functions and markers. Acta Biomater 2016; 43:3-13. [PMID: 27395828 DOI: 10.1016/j.actbio.2016.07.003] [Citation(s) in RCA: 229] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 06/08/2016] [Accepted: 07/05/2016] [Indexed: 02/07/2023]
Abstract
UNLABELLED The foreign body reaction (FBR) is a response of the host tissue against more or less degradation-resistant foreign macromolecular material. The reaction is divided into five different phases which involve most aspects of the innate and the adaptive immune system: protein adsorption, acute and chronic inflammation, foreign body giant cell formation and fibrosis. It is long known, that macrophages play a central role in all of these phases except for protein adsorption. Initially it was believed that the macrophage driven FBR has a complete negative effect on biocompatibility. Recent progress in biomaterial and macrophage research however describe macrophages as more than pure antigen phagocytosing and presenting cells and thus pro-inflammatory cells involved in biomaterial encapsulation and failure. Quite contrary, both, pro-inflammatory M1 macrophages, the diverse regulatory M2 macrophage subtypes and even foreign body giant cells (FBGC) are after necessary for integration of non-degradable biomaterials and degradation and replacement of degradable biomaterials. This review gives a comprehensive overview on the taxonomy of the currently known macrophage subtypes. Their diverging functions, metabolism and markers are summarized and the relevance of this more diverse macrophage picture for the design of biomaterials is shortly discussed. STATEMENT OF SIGNIFICANCE The view on role of macrophages in the foreign body reaction against biomaterials is rapidly changing. Despite the initial idea that macrophage are mainly involved in undesired degradation and biomaterial rejection it becomes now clear that they are nevertheless necessary for proper integration of non-degradable biomaterials and degradation of placeholder, degradable biomaterials. As a pathologist I experienced a lack on a good summary on the current taxonomy, functions and phenotypes of macrophages in my recent projects on the biocompatibility of biomaterials in the mouse model. The submitted review therefore intends to gives a comprehensive overview on the taxonomy of the currently known macrophage subtypes. Their diverging functions, metabolism and markers are summarized and the relevance of this more diverse macrophage picture for the design of biomaterials is shortly discussed.
Collapse
Affiliation(s)
- R Klopfleisch
- Institute of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Straße 15, Berlin 14163, Germany.
| |
Collapse
|
249
|
Das S, Majid M, Baker AB. Syndecan-4 enhances PDGF-BB activity in diabetic wound healing. Acta Biomater 2016; 42:56-65. [PMID: 27381525 DOI: 10.1016/j.actbio.2016.07.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 06/28/2016] [Accepted: 07/01/2016] [Indexed: 01/13/2023]
Abstract
UNLABELLED Non-healing ulcers are a common consequence of long-term diabetes and severe peripheral vascular disease. These non-healing wounds are a major source of morbidity in patients with diabetes and place a heavy financial burden on the healthcare system. Growth factor therapies are an attractive strategy for enhancing wound closure in non-healing wounds but have only achieved mixed results in clinical trials. Platelet derived growth factor-BB (PDGF-BB) is the only currently approved growth factor therapy for non-healing wounds. However, PDGF-BB therapy is not effective in many patients and requires high doses that increase the potential for side effects. In this work, we demonstrate that syndecan-4 delivered in a proteoliposomal formulation enhances PDGF-BB activity in diabetic wound healing. In particular, syndecan-4 proteoliposomes enhance the migration of keratinocytes derived from patients with diabetes. In addition, syndecan-4 proteoliposomes sensitize keratinocytes to PDGF-BB stimulation, enhancing the intracellular signaling response to PDGF-BB. We further demonstrated that co-therapy with syndecan-4 proteoliposomes enhanced wound closure in diabetic, hyperlipidemic ob/ob mice. Wounds treated with both syndecan-4 proteoliposomes and PDGF-BB had increased re-epithelization and angiogenesis in comparison to wounds treated with PDGF-BB alone. Moreover, the wounds treated with syndecan-4 proteoliposomes and PDGF-BB also had increased M2 macrophages and reduced M1 macrophages, suggesting syndecan-4 delivery induces immunomodulation within the healing wounds. Together our findings support that syndecan-4 proteoliposomes markedly improve PDGF-BB efficacy for wound healing and may be useful in enhancing treatments for non-healing wounds. STATEMENT OF SIGNIFICANCE Non-healing wounds are major healthcare issue for patients with diabetes and peripheral vascular disease. Growth factor therapies have potential for healing chronic wounds but have not been effective for many patients. PDGF-BB is currently the only approved growth factor for enhancing wound healing. However, it has not seen widespread adoption due to limited efficacy and high cost. In this work, we have developed an enhancing agent that improves the activity of PDGF-BB in promoting wound healing in animals with diabetes. This co-therapy may be useful in improving the efficacy of PDGFBB and enhance its safety through lowering the dose of growth factor needed to improve wound healing.
Collapse
Affiliation(s)
- Subhamoy Das
- Department of Biomedical Engineering, University of Texas, Austin, TX, United States
| | - Marjan Majid
- Department of Biomedical Engineering, University of Texas, Austin, TX, United States
| | - Aaron B Baker
- Department of Biomedical Engineering, University of Texas, Austin, TX, United States; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, United States; Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, United States; Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX, United States.
| |
Collapse
|
250
|
Vijayavenkataraman S, Lu WF, Fuh JYH. 3D bioprinting of skin: a state-of-the-art review on modelling, materials, and processes. Biofabrication 2016; 8:032001. [DOI: 10.1088/1758-5090/8/3/032001] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|