1
|
Kobetitsch S, Gierlikowska B, Kunert O, Mazen AM, Raab P, Kretschmer N, Donolo C, Pirker T, Bauer R, Kiss AK, Pferschy-Wenzig EM. Salvadora persica leaves: phytochemical profile and in vitro-inhibitory activity on inflammatory mediators implicated in periodontal disease. PHARMACEUTICAL BIOLOGY 2024; 62:563-576. [PMID: 39021070 PMCID: PMC11259067 DOI: 10.1080/13880209.2024.2374801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 06/22/2024] [Indexed: 07/20/2024]
Abstract
CONTEXT Virtually all parts of Salvadora persica L. (Salvadoraceae) are used in traditional medicine. The twigs and leaves are used for oral health, but leaves are far less investigated. OBJECTIVE This study assesses the oral health-promoting potential of S. persica leaves with emphasis on anti-inflammatory and antiproliferative effects and provides an in depth-characterization of their metabolite profile. MATERIALS AND METHODS Hot-water and methanolic S. persica leaf extracts (1, 10, and 100 µg/mL) and their major constituents (5, 10, and 50 µM), were subjected to cellular assays on IL-8 and TNFα release in LPS-stimulated human neutrophils, NO-release in LPS/IFNγ stimulated mouse macrophages, and proliferation of HNO97 human tongue carcinoma cells. Metabolite profiling was performed by UHPLC-HRMS analysis. Major constituents were isolated and structurally elucidated. RESULTS AND DISCUSSION Both extracts showed pronounced anti-inflammatory activity in LPS-stimulated neutrophils. Major identified compound classes were flavonoid glycosides, the glucosinolate glucotropaeolin, phenyl- and benzylglycoside sulfates, and megastigmane glycosylsulfates, the latter ones identified for the first time in S. persica. Glucotropaeolin strongly inhibited the release of IL-8 and TNF-α (13.3 ± 2.0 and 22.7 ± 2.6% of the release of stimulated control cells at 50 µM), while some flavonoids and 3-(3'-O-sulfo-β-d-glucopyranosyloxy)-7,8-dihydro-β-ionone, a newly isolated megastigmane glycosylsulfate, were moderately active. Benzylisothiocyanate, which is likely formed from glucotropaeolin during traditional application of S. persica, showed considerable antiproliferative activity (IC50 in HNO97 cells: 10.19 ± 0.72 µM) besides strongly inhibiting IL-8 and TNFα release. CONCLUSIONS Glucotropaeolin and benzylisothiocyanate are likely implicated in the oral health-promoting effects of S. persica leaves. The chemistry and pharmacology of the newly identified megastigmane glycosylsulfates should be further evaluated.
Collapse
Affiliation(s)
- Sabine Kobetitsch
- Institute of Pharmaceutical Sciences, Pharmacognosy, University of Graz, Graz, Austria
| | - Barbara Gierlikowska
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Warsaw, Poland
| | - Olaf Kunert
- Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, University of Graz, Graz, Austria
| | - Ahmed M.A. Mazen
- Botany and Microbiology Department, Faculty of Science, Sohag University, Sohag, Egypt
| | - Pia Raab
- Institute of Pharmaceutical Sciences, Pharmacognosy, University of Graz, Graz, Austria
| | - Nadine Kretschmer
- Institute of Pharmaceutical Sciences, Pharmacognosy, University of Graz, Graz, Austria
| | - Carina Donolo
- Institute of Pharmaceutical Sciences, Pharmacognosy, University of Graz, Graz, Austria
| | - Teresa Pirker
- Institute of Pharmaceutical Sciences, Pharmacognosy, University of Graz, Graz, Austria
| | - Rudolf Bauer
- Institute of Pharmaceutical Sciences, Pharmacognosy, University of Graz, Graz, Austria
| | - Anna K. Kiss
- Chair and Department of Pharmaceutical Biology, Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
2
|
Chen P, Zhang Z, Sakai L, Xu Y, Wang S, Lee KE, Geng B, Kim J, Zhao B, Wang Q, Wen H, Chandler HL, Zhu H. Neutrophil pyroptosis regulates corneal wound healing and post-injury neovascularisation. Clin Transl Med 2024; 14:e1762. [PMID: 39496510 PMCID: PMC11534482 DOI: 10.1002/ctm2.1762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 11/06/2024] Open
Abstract
RATIONALE The cornea is a unique structure that maintains its clarity by remaining avascular. Corneal injuries can lead to neovascularisation (CNV) and fibrosis and are the third most common cause of blindness worldwide. OBJECTIVE Corneal injuries induce an immune cell infiltration to initiate reparative processes. However, inflammation caused by sustained immune cell infiltration is known to be detrimental and can delay the healing process. This study was designed to understand the potential role of neutrophil and epithelial cell crosstalk in post-injury CNV. METHODS AND RESULTS Western blotting and immunostaining assays demonstrated that neutrophils infiltrated corneas and underwent pyroptosis following acute alkali injury. In vivo studies showed that genetic ablation of Gasdermin D (GsdmD), a key effector of pyroptosis, enhanced corneal re-epithelialisation and suppressed post-injury CNV. In vitro co-culture experiments revealed that interleukin-1β (IL-1β) was released from pyroptotic neutrophils which suppressed migration of murine corneal epithelial cells. Real-time RT-PCR and immunostaining assays identified two factors, Wnt5a and soluble fms-like tyrosine kinase-1 (sflt-1), highly expressed in newly healed epithelial cells. sflt-1 is known to promote corneal avascularity. Bone marrow transplantation, antibody mediated neutrophil depletion, and pharmacological inhibition of pyroptosis promoted corneal wound healing and inhibited CNV in an in vivo murine corneal injury model. CONCLUSION Taken together, our study reveals the importance of neutrophil/epithelium crosstalk and neutrophil pyroptosis in response to corneal injuries. Inhibition of neutrophil pyroptosis may serve as a potential treatment to promote corneal healing without CNV. KEY POINTS Neutrophil pyroptosis delays re-epithelialization after corneal injury Compromised re-epithelialization promotes corneal neovascularization after injury Inhibition of post-injury pyroptosis could be an effective therapy to promote corneal wound healing.
Collapse
Affiliation(s)
- Peng Chen
- Department of SurgeryThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Zhentao Zhang
- Department of SurgeryThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Lilian Sakai
- Department of SurgeryThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Yanping Xu
- Department of SurgeryThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Shanzhi Wang
- College of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Kyung Eun Lee
- Department of SurgeryThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Bingchuan Geng
- Department of SurgeryThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Jongsoo Kim
- Department of SurgeryThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Bao Zhao
- Department of Microbial Infection and ImmunityThe Ohio State UniversityColumbusOhioUSA
| | - Qiang Wang
- Department of SurgeryThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Haitao Wen
- Department of Microbial Infection and ImmunityThe Ohio State UniversityColumbusOhioUSA
| | | | - Hua Zhu
- Department of SurgeryThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| |
Collapse
|
3
|
Clayton SM, Shafikhani SH, Soulika AM. Macrophage and Neutrophil Dysfunction in Diabetic Wounds. Adv Wound Care (New Rochelle) 2024; 13:463-484. [PMID: 38695109 PMCID: PMC11535468 DOI: 10.1089/wound.2023.0149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Significance: The incidence of diabetes continues to rise throughout the world in an alarming rate. Diabetic patients often develop diabetic foot ulcers (DFUs), many of which do not heal. Non-healing DFUs are a major cause of hospitalization, amputation, and increased morbidity. Understanding the underlying mechanisms of impaired healing in DFU is crucial for its management. Recent Advances: This review focuses on the recent advancements on macrophages and neutrophils in diabetic wounds and DFUs. In particular, we discuss diabetes-induced dysregulations and dysfunctions of macrophages and neutrophils. Critical Issues: It is well established that diabetic wounds are characterized by stalled inflammation that results in impaired healing. Recent findings in the field suggest that dysregulation of macrophages and neutrophils plays a critical role in impaired healing in DFUs. The delineation of mechanisms that restore macrophage and neutrophil function in diabetic wound healing is the focus of intense investigation. Future Directions: The breadth of recently generated knowledge on the activity of macrophages and neutrophils in diabetic wound healing is impressive. Experimental models have delineated pathways that hold promise for the treatment of diabetic wounds and DFUs. These pathways may be useful targets for further clinical investigation.
Collapse
Affiliation(s)
- Shannon M. Clayton
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, California, USA
| | - Sasha H. Shafikhani
- Department of Internal Medicine, Division of Hematology, Oncology and Cell Therapy, Rush University, Chicago, Illinois, USA
| | - Athena M. Soulika
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, California, USA
| |
Collapse
|
4
|
Davis SC, Gil J, Solis M. Nitric Oxide as an Efficient Antimicrobial Treatment for Second-Degree Burn Wounds. Mil Med 2024:usae402. [PMID: 39178132 DOI: 10.1093/milmed/usae402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 08/25/2024] Open
Abstract
INTRODUCTION Nitric oxide (NO) is a lipophilic gas with potent antimicrobial activity. Several in vitro and in vivo studies have demonstrated the broad-spectrum antimicrobial activity of NO-releasing compounds against bacteria, viruses, and parasites. The objective of this study was to assess the efficacy of topical NO formations with sustained release on microbial reduction in wounds. MATERIALS AND METHODS Swine was used as the preclinical animal model because of the similarities of porcine skin to human skin. Second-degree burn wounds were created in 3 pigs and then inoculated with Methicillin-resistant Staphylococcus aureus, Acinetobacter baumannii, or Candida albicans and covered with polyurethane film dressings to create biofilms. After 48 hours, wounds were then treated daily and then recovered for the bacterial burden assessments. Statistical analysis was performed using IBM SPSS statistics 27 using one-way ANOVA. RESULTS All treatments significantly reduced (P ≤ .05) the bacterial counts between assessment days 4 and 7. Wounds treated with the NVN4000 (1.8%) exhibited greater than 99.7% bacterial reduction on days 4 and 7. Significant differences (P ≤ .05) were observed in wounds treated with NVN4000 (1.8%) compared to silver sulfadiazine. CONCLUSIONS These studies demonstrate that topical NO-releasing formulations effectively reduce the microbial burden of several microorganisms and exhibit superior antimicrobial efficacy compared to silver sulfadiazine in the porcine wound model.
Collapse
Affiliation(s)
- Stephen C Davis
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joel Gil
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Michael Solis
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
5
|
Zhang Y, Lu Q. Immune cells in skin inflammation, wound healing, and skin cancer. J Leukoc Biol 2024; 115:852-865. [PMID: 37718697 DOI: 10.1093/jleuko/qiad107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/22/2023] [Accepted: 09/05/2023] [Indexed: 09/19/2023] Open
Abstract
Given the self-evident importance of cutaneous immunity in the maintenance of body-surface homeostasis, disturbance of the steady-state skin is inextricably intertwined with dysfunction in cutaneous immunity. It is often overlooked by people that skin, well-known as a solid physical barrier, is also a strong immunological barrier, considering the abundant presence of immune cells including lymphocytes, granulocytes, dendritic cells, and macrophages. What's more, humoral immune components including cytokines, immunoglobulins, and antimicrobial peptides are also rich in the skin. This review centers on skin inflammation (acute and chronic, infection and aseptic inflammation), wound healing, and skin cancer to elucidate the elaborate network of immune cells in skin diseases.
Collapse
Affiliation(s)
- Yuhan Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
| |
Collapse
|
6
|
Cioce A, Cavani A, Cattani C, Scopelliti F. Role of the Skin Immune System in Wound Healing. Cells 2024; 13:624. [PMID: 38607063 PMCID: PMC11011555 DOI: 10.3390/cells13070624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/21/2024] [Accepted: 03/31/2024] [Indexed: 04/13/2024] Open
Abstract
Wound healing is a dynamic and complex process, characterized by the coordinated activities of multiple cell types, each with distinct roles in the stages of hemostasis, inflammation, proliferation, and remodeling. The cells of the immune system not only act as sentinels to monitor the skin and promote homeostasis, but they also play an important role in the process of skin wound repair. Skin-resident and recruited immune cells release cytokines and growth factors that promote the amplification of the inflammatory process. They also work with non-immune cells to remove invading pathogens and debris, as well as guide the regeneration of damaged host tissues. Dysregulation of the immune system at any stage of the process may lead to a prolongation of the inflammatory phase and the development of a pathological condition, such as a chronic wound. The present review aims to summarize the roles of different immune cells, with special emphasis on the different stages of the wound healing process.
Collapse
Affiliation(s)
| | | | | | - Fernanda Scopelliti
- National Institute for Health, Migration and Poverty INMP/NIHMP, Via di S.Gallicano, 25, 00153 Rome, Italy; (A.C.); (A.C.); (C.C.)
| |
Collapse
|
7
|
Metzemaekers M, Malengier-Devlies B, Gouwy M, De Somer L, Cunha FDQ, Opdenakker G, Proost P. Fast and furious: The neutrophil and its armamentarium in health and disease. Med Res Rev 2023; 43:1537-1606. [PMID: 37036061 DOI: 10.1002/med.21958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 12/27/2022] [Accepted: 03/24/2023] [Indexed: 04/11/2023]
Abstract
Neutrophils are powerful effector cells leading the first wave of acute host-protective responses. These innate leukocytes are endowed with oxidative and nonoxidative defence mechanisms, and play well-established roles in fighting invading pathogens. With microbicidal weaponry largely devoid of specificity and an all-too-well recognized toxicity potential, collateral damage may occur in neutrophil-rich diseases. However, emerging evidence suggests that neutrophils are more versatile, heterogeneous, and sophisticated cells than initially thought. At the crossroads of innate and adaptive immunity, neutrophils demonstrate their multifaceted functions in infectious and noninfectious pathologies including cancer, autoinflammation, and autoimmune diseases. Here, we discuss the kinetics of neutrophils and their products of activation from bench to bedside during health and disease, and provide an overview of the versatile functions of neutrophils as key modulators of immune responses and physiological processes. We focus specifically on those activities and concepts that have been validated with primary human cells.
Collapse
Affiliation(s)
- Mieke Metzemaekers
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Bert Malengier-Devlies
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Lien De Somer
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
- Division of Pediatric Rheumatology, University Hospital Leuven, Leuven, Belgium
- European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) at the University Hospital Leuven, Leuven, Belgium
| | | | - Ghislain Opdenakker
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Sousa AB, Barbosa JN. The Role of Neutrophils in Biomaterial-Based Tissue Repair-Shifting Paradigms. J Funct Biomater 2023; 14:327. [PMID: 37367291 DOI: 10.3390/jfb14060327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023] Open
Abstract
Tissue engineering and regenerative medicine are pursuing clinical valid solutions to repair and restore function of damaged tissues or organs. This can be achieved in different ways, either by promoting endogenous tissue repair or by using biomaterials or medical devices to replace damaged tissues. The understanding of the interactions of the immune system with biomaterials and how immune cells participate in the process of wound healing are critical for the development of successful solutions. Until recently, it was thought that neutrophils participate only in the initial steps of an acute inflammatory response with the role of eliminating pathogenic agents. However, the appreciation that upon activation the longevity of neutrophils is highly increased and the fact that neutrophils are highly plastic cells and can polarize into different phenotypes led to the discovery of new and important actions of neutrophils. In this review, we focus on the roles of neutrophils in the resolution of the inflammatory response, in biomaterial-tissue integration and in the subsequent tissue repair/regeneration. We also discuss the potential of neutrophils for biomaterial-based immunomodulation.
Collapse
Affiliation(s)
- Ana Beatriz Sousa
- i3S-Instituto de Inovação e Investigação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-125 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Rua Alfredo Allen, 208, 4200-125 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Judite N Barbosa
- i3S-Instituto de Inovação e Investigação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-125 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Rua Alfredo Allen, 208, 4200-125 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| |
Collapse
|
9
|
Jakovija A, Chtanova T. Skin immunity in wound healing and cancer. Front Immunol 2023; 14:1060258. [PMID: 37398649 PMCID: PMC10312005 DOI: 10.3389/fimmu.2023.1060258] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 05/24/2023] [Indexed: 07/04/2023] Open
Abstract
The skin is the body's largest organ. It serves as a barrier to pathogen entry and the first site of immune defense. In the event of a skin injury, a cascade of events including inflammation, new tissue formation and tissue remodeling contributes to wound repair. Skin-resident and recruited immune cells work together with non-immune cells to clear invading pathogens and debris, and guide the regeneration of damaged host tissues. Disruption to the wound repair process can lead to chronic inflammation and non-healing wounds. This, in turn, can promote skin tumorigenesis. Tumors appropriate the wound healing response as a way of enhancing their survival and growth. Here we review the role of resident and skin-infiltrating immune cells in wound repair and discuss their functions in regulating both inflammation and development of skin cancers.
Collapse
Affiliation(s)
- Arnolda Jakovija
- Immunity Theme, Garvan Institute of Medical Research, Sydney, Australia
- St. Vincent’s School of Medicine, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Tatyana Chtanova
- Immunity Theme, Garvan Institute of Medical Research, Sydney, Australia
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, Australia
| |
Collapse
|
10
|
Kidzeru EB, Lebeko M, Sharma JR, Nkengazong L, Adeola HA, Ndlovu H, P Khumalo N, Bayat A. Immune cells and associated molecular markers in dermal fibrosis with focus on raised cutaneous scars. Exp Dermatol 2023; 32:570-587. [PMID: 36562321 PMCID: PMC10947010 DOI: 10.1111/exd.14734] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/04/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Raised dermal scars including hypertrophic, and keloid scars as well as scalp-associated fibrosing Folliculitis Keloidalis Nuchae (FKN) are a group of fibrotic raised dermal lesions that mostly occur following cutaneous injury. They are characterized by increased extracellular matrix (ECM) deposition, primarily excessive collagen type 1 production by hyperproliferative fibroblasts. The extent of ECM deposition is thought to be proportional to the severity of local skin inflammation leading to excessive fibrosis of the dermis. Due to a lack of suitable study models, therapy for raised dermal scars remains ill-defined. Immune cells and their associated markers have been strongly associated with dermal fibrosis. Therefore, modulation of the immune system and use of anti-inflammatory cytokines are of potential interest in the management of dermal fibrosis. In this review, we will discuss the importance of immune factors in the pathogenesis of raised dermal scarring. The aim here is to provide an up-to-date comprehensive review of the literature, from PubMed, Scopus, and other relevant search engines in order to describe the known immunological factors associated with raised dermal scarring. The importance of immune cells including mast cells, macrophages, lymphocytes, and relevant molecules such as cytokines, chemokines, and growth factors, antibodies, transcription factors, and other immune-associated molecules as well as tissue lymphoid aggregates identified within raised dermal scars will be presented. A growing body of evidence points to a shift from proinflammatory Th1 response to regulatory/anti-inflammatory Th2 response being associated with the development of fibrogenesis in raised dermal scarring. In summary, a better understanding of immune cells and associated molecular markers in dermal fibrosis will likely enable future development of potential immune-modulated therapeutic, diagnostic, and theranostic targets in raised dermal scarring.
Collapse
Affiliation(s)
- Elvis Banboye Kidzeru
- Wound Healing And Keloid Scar Unit, Medical Research Council (South Africa), Hair and Skin Research Laboratory, Division of Dermatology, Department of MedicineUniversity of Cape TownCape TownSouth Africa
- Microbiology, Infectious Diseases, and Immunology Laboratory (LAMMII)Centre for Research on Health and Priority Pathologies (CRSPP)Institute of Medical Research and Medicinal Plant Studies (IMPM), Ministry of Scientific Research and InnovationYaoundéCameroon
| | - Maribanyana Lebeko
- Wound Healing And Keloid Scar Unit, Medical Research Council (South Africa), Hair and Skin Research Laboratory, Division of Dermatology, Department of MedicineUniversity of Cape TownCape TownSouth Africa
- Present address:
Cape Biologix Technologies (PTY, LTD)Cape TownSouth Africa
| | - Jyoti Rajan Sharma
- Wound Healing And Keloid Scar Unit, Medical Research Council (South Africa), Hair and Skin Research Laboratory, Division of Dermatology, Department of MedicineUniversity of Cape TownCape TownSouth Africa
- Biomedical Research and Innovation Platform, South African Medical Research Council, Francie van Zijl Drive, Parow ValleyCape TownSouth Africa
- Present address:
Biomedical Research and Innovation Platform, South African Medical Research Council, Francie van Zijl Drive, Parow ValleyCape TownSouth Africa
| | - Lucia Nkengazong
- Microbiology, Infectious Diseases, and Immunology Laboratory (LAMMII)Centre for Research on Health and Priority Pathologies (CRSPP)Institute of Medical Research and Medicinal Plant Studies (IMPM), Ministry of Scientific Research and InnovationYaoundéCameroon
| | - Henry Ademola Adeola
- Wound Healing And Keloid Scar Unit, Medical Research Council (South Africa), Hair and Skin Research Laboratory, Division of Dermatology, Department of MedicineUniversity of Cape TownCape TownSouth Africa
| | - Hlumani Ndlovu
- Department of Integrative Biomedical SciencesUniversity of Cape TownCape TownSouth Africa
| | - Nonhlanhla P Khumalo
- Wound Healing And Keloid Scar Unit, Medical Research Council (South Africa), Hair and Skin Research Laboratory, Division of Dermatology, Department of MedicineUniversity of Cape TownCape TownSouth Africa
| | - Ardeshir Bayat
- Wound Healing And Keloid Scar Unit, Medical Research Council (South Africa), Hair and Skin Research Laboratory, Division of Dermatology, Department of MedicineUniversity of Cape TownCape TownSouth Africa
| |
Collapse
|
11
|
Abstract
The epithelial tissues that line our body, such as the skin and gut, have remarkable regenerative prowess and continually renew throughout our lifetimes. Owing to their barrier function, these tissues have also evolved sophisticated repair mechanisms to swiftly heal and limit the penetration of harmful agents following injury. Researchers now appreciate that epithelial regeneration and repair are not autonomous processes but rely on a dynamic cross talk with immunity. A wealth of clinical and experimental data point to the functional coupling of reparative and inflammatory responses as two sides of the same coin. Here we bring to the fore the immunological signals that underlie homeostatic epithelial regeneration and restitution following damage. We review our current understanding of how immune cells contribute to distinct phases of repair. When unchecked, immune-mediated repair programs are co-opted to fuel epithelial pathologies such as cancer, psoriasis, and inflammatory bowel diseases. Thus, understanding the reparative functions of immunity may advance therapeutic innovation in regenerative medicine and epithelial inflammatory diseases.
Collapse
Affiliation(s)
- Laure Guenin-Mace
- Department of Pathology, NYU Langone Health, New York, NY, USA;
- Immunobiology and Therapy Unit, INSERM U1224, Institut Pasteur, Paris, France
| | - Piotr Konieczny
- Department of Pathology, NYU Langone Health, New York, NY, USA;
| | - Shruti Naik
- Department of Pathology, NYU Langone Health, New York, NY, USA;
- Department of Medicine, Ronald O. Perelman Department of Dermatology, and Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| |
Collapse
|
12
|
Lei XX, Zou CY, Hu JJ, Jiang YL, Zhang XZ, Zhao LM, He T, Zhang QY, Li YX, Li-Ling J, Xie HQ. Click-crosslinked in-situ hydrogel improves the therapeutic effect in wound infections through antibacterial, antioxidant and anti-inflammatory activities. CHEMICAL ENGINEERING JOURNAL 2023; 461:142092. [DOI: 10.1016/j.cej.2023.142092] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2024]
|
13
|
Siwicki M, Kubes P. Neutrophils in host defense, healing, and hypersensitivity: Dynamic cells within a dynamic host. J Allergy Clin Immunol 2023; 151:634-655. [PMID: 36642653 DOI: 10.1016/j.jaci.2022.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 11/11/2022] [Accepted: 12/02/2022] [Indexed: 01/15/2023]
Abstract
Neutrophils are cells of the innate immune system that are extremely abundant in vivo and respond quickly to infection, injury, and inflammation. Their constant circulation throughout the body makes them some of the first responders to infection, and indeed they play a critical role in host defense against bacterial and fungal pathogens. It is now appreciated that neutrophils also play an important role in tissue healing after injury. Their short life cycle, rapid response kinetics, and vast numbers make neutrophils a highly dynamic and potentially extremely influential cell population. It has become clear that they are highly integrated with other cells of the immune system and can thus exert critical effects on the course of an inflammatory response; they can further impact tissue homeostasis and recovery after challenge. In this review, we discuss the fundamentals of neutrophils in host defense and healing; we explore the relationship between neutrophils and the dynamic host environment, including circadian cycles and the microbiome; we survey the field of neutrophils in asthma and allergy; and we consider the question of neutrophil heterogeneity-namely, whether there could be specific subsets of neutrophils that perform different functions in vivo.
Collapse
Affiliation(s)
- Marie Siwicki
- Immunology Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Paul Kubes
- Immunology Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
14
|
Abstract
Coronavirus disease 2019 (COVID-19) is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and has resulted in more than 6 million deaths worldwide. COVID-19 is a respiratory disease characterized by pulmonary dysfunction leading to acute respiratory distress syndrome (ARDs), as well as disseminated coagulation, and multi-organ dysfunction. Neutrophils and neutrophil extracellular traps (NETs) have been implicated in the pathogenesis of COVID-19. In this review, we highlight key gaps in knowledge, discuss the heterogeneity of neutrophils during the evolution of the disease, how they can contribute to COVID-19 pathogenesis, and potential therapeutic strategies that target neutrophil-mediated inflammatory responses.
Collapse
Affiliation(s)
- Fernanda V. S. Castanheira
- Department of Physiology and PharmacologyUniversity of CalgaryCalgaryAlbertaCanada
- Department of Microbiology, Immunology and InfectiousUniversity of CalgaryCalgaryAlbertaCanada
- Snyder Institute for Chronic DiseasesUniversity of CalgaryCalgaryAlbertaCanada
| | - Paul Kubes
- Department of Physiology and PharmacologyUniversity of CalgaryCalgaryAlbertaCanada
- Department of Microbiology, Immunology and InfectiousUniversity of CalgaryCalgaryAlbertaCanada
- Snyder Institute for Chronic DiseasesUniversity of CalgaryCalgaryAlbertaCanada
| |
Collapse
|
15
|
Putative Role of Neutrophil Extracellular Trap Formation in Chronic Myeloproliferative Neoplasms. Int J Mol Sci 2023; 24:ijms24054497. [PMID: 36901933 PMCID: PMC10003516 DOI: 10.3390/ijms24054497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 03/03/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs) are hematologic malignancies characterized by gene mutations that promote myeloproliferation and resistance to apoptosis via constitutively active signaling pathways, with Janus kinase 2-signal transducers and the activators of transcription (JAK-STAT) axis as a core part. Chronic inflammation has been described as a pivot for the development and advancement of MPNs from early stage cancer to pronounced bone marrow fibrosis, but there are still unresolved questions regarding this issue. The MPN neutrophils are characterized by upregulation of JAK target genes, they are in a state of activation and with deregulated apoptotic machinery. Deregulated neutrophil apoptotic cell death supports inflammation and steers them towards secondary necrosis or neutrophil extracellular trap (NET) formation, a trigger of inflammation both ways. NETs in proinflammatory bone marrow microenvironment induce hematopoietic precursor proliferation, which has an impact on hematopoietic disorders. In MPNs, neutrophils are primed for NET formation, and even though it seems obvious for NETs to intervene in the disease progression by supporting inflammation, no reliable data are available. We discuss in this review the potential pathophysiological relevance of NET formation in MPNs, with the intention of contributing to a better understanding of how neutrophils and neutrophil clonality can orchestrate the evolution of a pathological microenvironment in MPNs.
Collapse
|
16
|
Sansores-España LD, Melgar-Rodríguez S, Vernal R, Carrillo-Ávila BA, Martínez-Aguilar VM, Díaz-Zúñiga J. Neutrophil N1 and N2 Subsets and Their Possible Association with Periodontitis: A Scoping Review. Int J Mol Sci 2022; 23:12068. [PMID: 36292925 PMCID: PMC9603394 DOI: 10.3390/ijms232012068] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/02/2022] [Accepted: 10/06/2022] [Indexed: 12/04/2022] Open
Abstract
Periodontitis is a chronic non-communicable disease caused by dysbiotic changes that affect the subgingival microbiota. During periodontitis, neutrophils play a central role in the initial recognition of bacteria, and their number increases with the appearance of the first signs of periodontal inflammation. Recent evidence has led to the proposition that neutrophils can also functionally polarize, determining selective activity patterns related to different diseases. Two well-defined neutrophil phenotypes have been described, the pro-inflammatory N1 subset and the suppressor N2 subset. To date, it has not been established whether these different neutrophil subtypes play a role in the pathogenesis of periodontitis. Thus, this scoping review aimed to determine whether there was evidence to suggest that the neutrophils present in periodontal tissues can be associated with certain phenotypes. The research question, population, concept, and context sought to identify original articles, in humans, that detected the presence of neutrophils in the periodontal tissues of people affected by periodontitis. Based on the search strategy, we found 3658 studies. After removing the papers with abstracts not related to the outcome measures and eligibility criteria, 16 articles were included for qualitative analysis. Several studies identified the presence of different neutrophil subsets, specifically, the naive, pro- and para-inflammatory, hyper-reactive and hyper-active, and high- and low-responder phenotypes. The existing evidence demonstrates the presence of pro-inflammatory, hyper-reactive and high-responder neutrophils in periodontal tissues affected with periodontitis. There is no evidence demonstrating the presence of the N1 or N2 phenotypes in periodontal tissues during periodontitis. However, the existence of pro-inflammatory phenotypes, which increase NETosis and degranulation, and increase the production of pro-inflammatory cytokines, could be suggestive of the N1 phenotypes.
Collapse
Affiliation(s)
- Luis Daniel Sansores-España
- Faculty of Dentistry, Autonomous University of Yucatán, Merida 97000, Mexico
- Periodontal Biology Laboratory, Faculty of Dentistry, University of Chile, Santiago 8380492, Chile
| | - Samanta Melgar-Rodríguez
- Periodontal Biology Laboratory, Faculty of Dentistry, University of Chile, Santiago 8380492, Chile
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, University of Chile, Santiago 8380492, Chile
| | | | | | - Jaime Díaz-Zúñiga
- Periodontal Biology Laboratory, Faculty of Dentistry, University of Chile, Santiago 8380492, Chile
- Department of Medicine, Faculty of Medicine, University of Atacama, Copiapo 7500015, Chile
| |
Collapse
|
17
|
Modeling HPV-Associated Disease and Cancer Using the Cottontail Rabbit Papillomavirus. Viruses 2022; 14:v14091964. [PMID: 36146770 PMCID: PMC9503101 DOI: 10.3390/v14091964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 01/06/2023] Open
Abstract
Approximately 5% of all human cancers are attributable to human papillomavirus (HPV) infections. HPV-associated diseases and cancers remain a substantial public health and economic burden worldwide despite the availability of prophylactic HPV vaccines. Current diagnosis and treatments for HPV-associated diseases and cancers are predominantly based on cell/tissue morphological examination and/or testing for the presence of high-risk HPV types. There is a lack of robust targets/markers to improve the accuracy of diagnosis and treatments. Several naturally occurring animal papillomavirus models have been established as surrogates to study HPV pathogenesis. Among them, the Cottontail rabbit papillomavirus (CRPV) model has become known as the gold standard. This model has played a pivotal role in the successful development of vaccines now available to prevent HPV infections. Over the past eighty years, the CRPV model has been widely applied to study HPV carcinogenesis. Taking advantage of a large panel of functional mutant CRPV genomes with distinct, reproducible, and predictable phenotypes, we have gained a deeper understanding of viral–host interaction during tumor progression. In recent years, the application of genome-wide RNA-seq analysis to the CRPV model has allowed us to learn and validate changes that parallel those reported in HPV-associated cancers. In addition, we have established a selection of gene-modified rabbit lines to facilitate mechanistic studies and the development of novel therapeutic strategies. In the current review, we summarize some significant findings that have advanced our understanding of HPV pathogenesis and highlight the implication of the development of novel gene-modified rabbits to future mechanistic studies.
Collapse
|
18
|
Montelukast, an Antagonist of Cysteinyl Leukotriene Signaling, Impairs Burn Wound Healing. Plast Reconstr Surg 2022; 150:92e-104e. [PMID: 35536768 DOI: 10.1097/prs.0000000000009228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Burns are severe injuries often associated with impaired wound healing. Impaired healing is caused by multiple factors, including dysregulated inflammatory responses at the wound site. Interestingly, montelukast, an antagonist for cysteinyl leukotrienes and U.S. Food and Drug Administration approved for treatment of asthma and allergy, was previously shown to enhance healing in excision wounds and to modulate local inflammation. METHODS In this study, the authors examined the effect of montelukast on wound healing in a mouse model of scald burn injury. Burn wound tissues isolated from montelukast- and vehicle-treated mice at various times after burn injury were analyzed for wound areas ( n = 34 to 36), reepithelialization ( n = 14), inflammation ( n = 8 to 9), and immune cell infiltration ( n = 3 to 6) and proliferation ( n = 7 to 8). RESULTS In contrast to previously described beneficial effects in excision wounds, this study shows that montelukast delays burn wound healing by impairing the proliferation of keratinocytes and endothelial cells. This occurs largely independently of inflammatory responses at the wound site, suggesting that montelukast impairs specifically the proliferative phase of wound healing in burns. Wound healing rates in mice in which leukotrienes are not produced were not affected by montelukast. CONCLUSION Montelukast delays wound healing mainly by reducing the proliferation of local cells after burn injury. CLINICAL RELEVANCE STATEMENT Although additional and clinical studies are necessary, our study suggests that burn patients who are on montelukast may exhibit delayed healing, necessitating extra observation.
Collapse
|
19
|
Muire PJ, Thompson MA, Christy RJ, Natesan S. Advances in Immunomodulation and Immune Engineering Approaches to Improve Healing of Extremity Wounds. Int J Mol Sci 2022; 23:4074. [PMID: 35456892 PMCID: PMC9032453 DOI: 10.3390/ijms23084074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/01/2022] [Accepted: 04/03/2022] [Indexed: 12/04/2022] Open
Abstract
Delayed healing of traumatic wounds often stems from a dysregulated immune response initiated or exacerbated by existing comorbidities, multiple tissue injury or wound contamination. Over decades, approaches towards alleviating wound inflammation have been centered on interventions capable of a collective dampening of various inflammatory factors and/or cells. However, a progressive understanding of immune physiology has rendered deeper knowledge on the dynamic interplay of secreted factors and effector cells following an acute injury. There is a wide body of literature, both in vitro and in vivo, abstracted on the immunomodulatory approaches to control inflammation. Recently, targeted modulation of the immune response via biotechnological approaches and biomaterials has gained attention as a means to restore the pro-healing phenotype and promote tissue regeneration. In order to fully realize the potential of these approaches in traumatic wounds, a critical and nuanced understanding of the relationships between immune dysregulation and healing outcomes is needed. This review provides an insight on paradigm shift towards interventional approaches to control exacerbated immune response following a traumatic injury from an agonistic to a targeted path. We address such a need by (1) providing a targeted discussion of the wound healing processes to assist in the identification of novel therapeutic targets and (2) highlighting emerging technologies and interventions that utilize an immunoengineering-based approach. In addition, we have underscored the importance of immune engineering as an emerging tool to provide precision medicine as an option to modulate acute immune response following a traumatic injury. Finally, an overview is provided on how an intervention can follow through a successful clinical application and regulatory pathway following laboratory and animal model evaluation.
Collapse
Affiliation(s)
- Preeti J. Muire
- Combat Wound Care Research Department, US Army Institute of Surgical Research, JBSA Ft Sam Houston, San Antonio, TX 78234, USA; (M.A.T.); (R.J.C.)
| | | | | | - Shanmugasundaram Natesan
- Combat Wound Care Research Department, US Army Institute of Surgical Research, JBSA Ft Sam Houston, San Antonio, TX 78234, USA; (M.A.T.); (R.J.C.)
| |
Collapse
|
20
|
Kolesnikoff N, Chen CH, Samuel M. Interrelationships between the extracellular matrix and the immune microenvironment that govern epithelial tumour progression. Clin Sci (Lond) 2022; 136:361-377. [PMID: 35260891 PMCID: PMC8907655 DOI: 10.1042/cs20210679] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/18/2022] [Accepted: 02/24/2022] [Indexed: 12/19/2022]
Abstract
Solid tumours are composed of cancer cells characterised by genetic mutations that underpin the disease, but also contain a suite of genetically normal cells and the extracellular matrix (ECM). These two latter components are constituents of the tumour microenvironment (TME), and are key determinants of tumour biology and thereby the outcomes for patients. The tumour ECM has been the subject of intense research over the past two decades, revealing key biochemical and mechanobiological principles that underpin its role in tumour cell proliferation and survival. However, the ECM also strongly influences the genetically normal immune cells within the microenvironment, regulating not only their proliferation and survival, but also their differentiation and access to tumour cells. Here we review recent advances in our knowledge of how the ECM regulates the tumour immune microenvironment and vice versa, comparing normal skin wound healing to the pathological condition of tumour progression.
Collapse
Affiliation(s)
- Natasha Kolesnikoff
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - Chun-Hsien Chen
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - Michael Susithiran Samuel
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
- Adelaide Medical School, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
21
|
Syahirah R, Hsu AY, Deng Q. A curious case of cyclin‐dependent kinases in neutrophils. J Leukoc Biol 2022; 111:1057-1068. [PMID: 35188696 PMCID: PMC9035055 DOI: 10.1002/jlb.2ru1021-573r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/21/2022] [Accepted: 01/31/2022] [Indexed: 12/11/2022] Open
Abstract
Neutrophils are terminally differentiated, short-lived white blood cells critical for innate immunity. Although cyclin-dependent kinases (CDKs) are typically related to cell cycle progression, increasing evidence has shown that they regulate essential functions of neutrophils. This review highlights the roles of CDKs and their partners, cyclins, in neutrophils, outside of cell cycle regulation. CDK1-10 and several cyclins are expressed in neutrophils, albeit at different levels. Observed phenotypes associated with specific inhibition or genetic loss of CDK2 indicate its role in modulating neutrophil migration. CDK4 and 6 regulate neutrophil extracellular traps (NETs) formation, while CDK5 regulates neutrophil degranulation. CDK7 and 9 are critical in neutrophil apoptosis, contributing to inflammation resolution. In addition to the CDKs that regulate mature neutrophil functions, cyclins are essential in hematopoiesis and granulopoiesis. The pivotal roles of CDKs in neutrophils present an untapped potential in targeting CDKs for treating neutrophil-dominant inflammatory diseases and understanding the regulation of the neutrophil life cycle.
Collapse
Affiliation(s)
- Ramizah Syahirah
- Department of Biological Sciences Purdue University West Lafayette Indiana USA
| | - Alan Y. Hsu
- Department of Biological Sciences Purdue University West Lafayette Indiana USA
- Department of Pathology Harvard Medical School Boston Massachusetts USA
- Department of Laboratory Medicine The Stem Cell Program, Boston Children's Hospital Boston Massachusetts USA
| | - Qing Deng
- Department of Biological Sciences Purdue University West Lafayette Indiana USA
- Purdue Institute of Inflammation Immunology and Infectious Disease, Purdue University West Lafayette Indiana USA
- Purdue University Center for Cancer Research, Purdue University West Lafayette Indiana USA
| |
Collapse
|
22
|
Dubey R, Prabhakar PK, Gupta J. Epigenetics: key to improve delayed wound healing in type 2 diabetes. Mol Cell Biochem 2022; 477:371-383. [PMID: 34739665 DOI: 10.1007/s11010-021-04285-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/23/2021] [Indexed: 12/13/2022]
Abstract
Diabetes-related delayed wound healing is a multifactorial, nuanced, and intertwined complication that causes substantial clinical morbidity. The etiology of diabetes and its related microvascular complications is affected by genes, diet, and lifestyle factors. Epigenetic modifications such as DNA methylation, histone modifications, and post-transcriptional RNA regulation (microRNAs) are subsequently recognized as key facilitators of the complicated interaction between genes and the environment. Current research suggests that diabetes-persuaded dysfunction of epigenetic pathways, which results in changed expression of genes in target cells and cause diabetes-related complications including cardiomyopathy, nephropathy, retinopathy, delayed wound healing, etc., which are foremost drivers to diabetes-related adverse outcomes. In this paper, we discuss the role of epigenetic mechanisms in controlling tissue repair, angiogenesis, and expression of growth factors, as well as recent findings that show the alteration of epigenetic events during diabetic wound healing.
Collapse
Affiliation(s)
- Rupal Dubey
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University (LPU), Jalandhar-Delhi G.T. Road, 144411, Phagwara, Punjab, India
| | - Pranav Kumar Prabhakar
- Department of Medical Laboratory Sciences, School of Physiotherapy and Paramedical Sciences, Lovely Professional University, 144411, Phagwara, Punjab, India
| | - Jeena Gupta
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University (LPU), Jalandhar-Delhi G.T. Road, 144411, Phagwara, Punjab, India.
| |
Collapse
|
23
|
Stephen B, Hajjar J. Immune System in Action. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1342:1-43. [PMID: 34972961 DOI: 10.1007/978-3-030-79308-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Tumor exists as a complex network of structures with an ability to evolve and evade the host immune surveillance mechanism. The immune milieu which includes macrophages, dendritic cells, natural killer cells, neutrophils, mast cells, B cells, and T cells is found in the core, the invasive margin, or the adjacent stromal or lymphoid component of the tumor. The immune infiltrate is heterogeneous and varies within a patient and between patients of the same tumor histology. The location, density, functionality, and the crosstalk between the immune cells in the tumor microenvironment influence the nature of immune response, prognosis, and treatment outcomes in cancer patients. Therefore, an understanding of the characteristics of the immune cells and their role in tumor immune surveillance is of paramount importance to identify immune targets and to develop novel immune therapeutics in the war against cancer. In this chapter, we provide an overview of the individual components of the human immune system and the translational relevance of predictive biomarkers.
Collapse
Affiliation(s)
- Bettzy Stephen
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Joud Hajjar
- Assistant Professor, Service Chief of Adult Allergy & Immunology, Division of Immunology, Allergy & Retrovirology, Baylor College of Medicine and Texas Children' Hospital, Houston, TX, USA
| |
Collapse
|
24
|
Hawthorne B, Simmons JK, Stuart B, Tung R, Zamierowski DS, Mellott AJ. Enhancing wound healing dressing development through interdisciplinary collaboration. J Biomed Mater Res B Appl Biomater 2021; 109:1967-1985. [PMID: 34002476 PMCID: PMC8519107 DOI: 10.1002/jbm.b.34861] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/03/2021] [Accepted: 04/30/2021] [Indexed: 12/31/2022]
Abstract
The process of wound healing includes four phases: Hemostasis, inflammation, proliferation, and remodeling. Many wound dressings and technologies have been developed to enhance the body's ability to close wounds and restore the function of damaged tissues. Several advancements in wound healing technology have resulted from innovative experiments by individual scientists or physicians working independently. The interplay between the medical and scientific research fields is vital to translating new discoveries in the lab to treatments at the bedside. Tracing the history of wound dressing development reveals that there is an opportunity for deeper collaboration between multiple disciplines to accelerate the advancement of novel wound healing technologies. In this review, we explore the different types of wound dressings and biomaterials used to treat wounds, and we investigate the role of multidisciplinary collaboration in the development of various wound management technologies to illustrate the benefit of direct collaboration between physicians and scientists.
Collapse
Affiliation(s)
- Briauna Hawthorne
- Department of Plastic SurgeryUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - J. Kai Simmons
- Department of Plastic SurgeryUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Braden Stuart
- Department of Plastic SurgeryUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Robert Tung
- Department of Plastic SurgeryUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - David S. Zamierowski
- Department of Plastic SurgeryUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Adam J. Mellott
- Department of Plastic SurgeryUniversity of Kansas Medical CenterKansas CityKansasUSA
| |
Collapse
|
25
|
Xu FW, Lv YL, Zhong YF, Xue YN, Wang Y, Zhang LY, Hu X, Tan WQ. Beneficial Effects of Green Tea EGCG on Skin Wound Healing: A Comprehensive Review. Molecules 2021; 26:6123. [PMID: 34684703 PMCID: PMC8540743 DOI: 10.3390/molecules26206123] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/18/2022] Open
Abstract
Epigallocatechin gallate (EGCG) is associated with various health benefits. In this review, we searched current work about the effects of EGCG and its wound dressings on skin for wound healing. Hydrogels, nanoparticles, micro/nanofiber networks and microneedles are the major types of EGCG-containing wound dressings. The beneficial effects of EGCG and its wound dressings at different stages of skin wound healing (hemostasis, inflammation, proliferation and tissue remodeling) were summarized based on the underlying mechanisms of antioxidant, anti-inflammatory, antimicrobial, angiogenesis and antifibrotic properties. This review expatiates on the rationale of using EGCG to promote skin wound healing and prevent scar formation, which provides a future clinical application direction of EGCG.
Collapse
Affiliation(s)
- Fa-Wei Xu
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, China; (F.-W.X.); (Y.-F.Z.); (Y.-N.X.); (Y.W.); (L.-Y.Z.); (X.H.)
| | - Ying-Li Lv
- Tea Research Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310013, China;
| | - Yu-Fan Zhong
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, China; (F.-W.X.); (Y.-F.Z.); (Y.-N.X.); (Y.W.); (L.-Y.Z.); (X.H.)
| | - Ya-Nan Xue
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, China; (F.-W.X.); (Y.-F.Z.); (Y.-N.X.); (Y.W.); (L.-Y.Z.); (X.H.)
| | - Yong Wang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, China; (F.-W.X.); (Y.-F.Z.); (Y.-N.X.); (Y.W.); (L.-Y.Z.); (X.H.)
| | - Li-Yun Zhang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, China; (F.-W.X.); (Y.-F.Z.); (Y.-N.X.); (Y.W.); (L.-Y.Z.); (X.H.)
| | - Xian Hu
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, China; (F.-W.X.); (Y.-F.Z.); (Y.-N.X.); (Y.W.); (L.-Y.Z.); (X.H.)
| | - Wei-Qiang Tan
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou 310016, China; (F.-W.X.); (Y.-F.Z.); (Y.-N.X.); (Y.W.); (L.-Y.Z.); (X.H.)
| |
Collapse
|
26
|
Nolan E, Malanchi I. Connecting the dots: Neutrophils at the interface of tissue regeneration and cancer. Semin Immunol 2021; 57:101598. [PMID: 35221216 PMCID: PMC9232712 DOI: 10.1016/j.smim.2022.101598] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/19/2022] [Accepted: 02/08/2022] [Indexed: 11/28/2022]
Abstract
Knowledge about neutrophil biology has exponentially grown over the past decades. A high volume of investigations focusing on the characterization of their initially unappreciated multifaceted functions have grown in parallel with the immunity and the cancer fields. This has led to a significant gain in knowledge about their functions not only in tissue defence against pathogens and the collateral damage their overactivation can cause, but also their role in tissue repair and regeneration especially in the context of sterile injuries. On the other hand, the cancer field has also intensively focused its attention on neutrophil engagement in the many steps of the tumorigenic process. This review aims to draw the readers' attention to the similar functions described for neutrophils in tissue repair and in cancer. By bridging the two fields, we provide support for the hypothesis that the underlying program driving cancer-dependent exploitation of neutrophils is rooted in their physiologic tissue protection functions. In this view, cross-fertilization between the two fields will expedite the discovery of therapeutic interventions based on neutrophil targeting or their manipulation.
Collapse
Affiliation(s)
- Emma Nolan
- Tumour Host Interaction Laboratory, The Francis Crick Institute, 1 Midland Road, NW1 1AT London, United Kingdom
| | - Ilaria Malanchi
- Tumour Host Interaction Laboratory, The Francis Crick Institute, 1 Midland Road, NW1 1AT London, United Kingdom.
| |
Collapse
|
27
|
Schmitt T, Katz N, Kishore V. A Feasibility Study on 3D Bioprinting of Microfat Constructs Towards Wound Healing Applications. Front Bioeng Biotechnol 2021; 9:707098. [PMID: 34386485 PMCID: PMC8353388 DOI: 10.3389/fbioe.2021.707098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/29/2021] [Indexed: 11/16/2022] Open
Abstract
Chronic wounds affect over 400,000 people in the United States alone, with up to 60,000 deaths each year from non-healing ulcerations. Tissue grafting (e.g., autografts, allografts, and xenografts) and synthetic skin substitutes are common treatment methods, but most solutions are limited to symptomatic treatment and do not address the underlying causes of the chronic wound. Use of fat grafts for wound healing applications has demonstrated promise but these grafts suffer from low cell viability and poor retention at the wound site resulting in suboptimal healing of chronic wounds. Herein, we report on an innovative closed-loop fat processing system (MiniTCTM) that can efficiently process lipoaspirates into microfat clusters comprising of highly viable regenerative cell population (i.e., adipose stromal cells, endothelial progenitors) preserved in their native niche. Cryopreservation of MiniTCTM isolated microfat retained cell count and viability. To improve microfat retention and engraftment at the wound site, microfat was mixed with methacrylated collagen (CMA) bioink and 3D printed to generate microfat-laden collagen constructs. Modulating the concentration of microfat in CMA constructs had no effect on print fidelity or stability of the printed constructs. Results from the Alamar blue assay showed that the cells remain viable and metabolically active in microfat-laden collagen constructs for up to 10 days in vitro. Further, quantitative assessment of cell culture medium over time using ELISA revealed a temporal expression of proinflammatory and anti-inflammatory cytokines indicative of wound healing microenvironment progression. Together, these results demonstrate that 3D bioprinting of microfat-laden collagen constructs is a promising approach to generate viable microfat grafts for potential use in treatment of non-healing chronic wounds.
Collapse
Affiliation(s)
- Trevor Schmitt
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, United States
| | - Nathan Katz
- Jointechlabs Inc., North Barrington, IL, United States
| | - Vipuil Kishore
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, United States
| |
Collapse
|
28
|
Wortel IMN, Niculescu I, Kolijn PM, Gov NS, de Boer RJ, Textor J. Local actin dynamics couple speed and persistence in a cellular Potts model of cell migration. Biophys J 2021; 120:2609-2622. [PMID: 34022237 PMCID: PMC8390880 DOI: 10.1016/j.bpj.2021.04.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 03/24/2021] [Accepted: 04/14/2021] [Indexed: 12/28/2022] Open
Abstract
Cell migration is astoundingly diverse. Molecular signatures, cell-cell interactions, and environmental structures each play their part in shaping cell motion, yielding numerous morphologies and migration modes. Nevertheless, in recent years, a simple unifying law was found to describe cell migration across many different cell types and contexts: faster cells turn less frequently. This universal coupling between speed and persistence (UCSP) was explained by retrograde actin flow from front to back, but it remains unclear how this mechanism generalizes to cells with complex shapes and cells migrating in structured environments, which may not have a well-defined front-to-back orientation. Here, we present an in-depth characterization of an existing cellular Potts model, in which cells polarize dynamically from a combination of local actin dynamics (stimulating protrusions) and global membrane tension along the perimeter (inhibiting protrusions). We first show that the UCSP emerges spontaneously in this model through a cross talk of intracellular mechanisms, cell shape, and environmental constraints, resembling the dynamic nature of cell migration in vivo. Importantly, we find that local protrusion dynamics suffice to reproduce the UCSP-even in cases in which no clear global, front-to-back polarity exists. We then harness the spatial nature of the cellular Potts model to show how cell shape dynamics limit both the speed and persistence a cell can reach and how a rigid environment such as the skin can restrict cell motility even further. Our results broaden the range of potential mechanisms underlying the speed-persistence coupling that has emerged as a fundamental property of migrating cells.
Collapse
Affiliation(s)
- Inge M N Wortel
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands; Data Science, Institute for Computing and Information Sciences, Radboud University, Nijmegen, the Netherlands.
| | - Ioana Niculescu
- Theoretical Biology and Bioinformatics, Department of Biology, Utrecht University, Utrecht, the Netherlands
| | - P Martijn Kolijn
- Theoretical Biology and Bioinformatics, Department of Biology, Utrecht University, Utrecht, the Netherlands
| | - Nir S Gov
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot, Israel
| | - Rob J de Boer
- Theoretical Biology and Bioinformatics, Department of Biology, Utrecht University, Utrecht, the Netherlands
| | - Johannes Textor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands; Data Science, Institute for Computing and Information Sciences, Radboud University, Nijmegen, the Netherlands.
| |
Collapse
|
29
|
Xu Z, Liang B, Tian J, Wu J. Anti-inflammation biomaterial platforms for chronic wound healing. Biomater Sci 2021; 9:4388-4409. [PMID: 34013915 DOI: 10.1039/d1bm00637a] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nowadays, there has been an increase in the number of people with chronic wounds, which has resulted in serious health problems worldwide. The rate-limiting stage of chronic wound healing has been found to be the inflammation stage, and strategies for shortening the prolonged inflammatory response have proven to be effective for increasing the healing rate. Recently, various anti-inflammatory strategies (such as anti-inflammatory drugs, antioxidant, NO regulation, antibacterial, immune regulation and angiogenesis) have attracted attention as potential therapeutic pathways. Moreover, various biomaterial platforms based on anti-inflammation therapy strategies have also emerged in the spotlight as potential therapies to accelerate the repair of chronic wounds. In this review, we systematically investigated the advances of various biomaterial platforms based on anti-inflammation strategies for chronic wound healing, to provide valuable guidance for future breakthroughs in chronic wound treatment.
Collapse
Affiliation(s)
- Zejun Xu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, P. R. China.
| | - Biao Liang
- Center of Digestive Endoscopy, Guangdong Second Provincial general Hospital, No. 466, Xingang Middle Road, Guangzhou 510317, Haizhu District, China.
| | - Junzhang Tian
- Center of Digestive Endoscopy, Guangdong Second Provincial general Hospital, No. 466, Xingang Middle Road, Guangzhou 510317, Haizhu District, China.
| | - Jun Wu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, P. R. China.
| |
Collapse
|
30
|
Shen P, Chen Y, Luo S, Fan Z, Wang J, Chang J, Deng J. Applications of biomaterials for immunosuppression in tissue repair and regeneration. Acta Biomater 2021; 126:31-44. [PMID: 33722787 DOI: 10.1016/j.actbio.2021.03.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/24/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022]
Abstract
The immune system plays an essential role in tissue repair and regeneration. Regardless of innate or adaptive immune responses, immunosuppressive strategies such as macrophage polarization and regulatory T (Treg) cell induction can be used to modulate the immune system to promote tissue repair and regeneration. Biomaterials can improve the production of anti-inflammatory macrophages and Treg cells by providing physiochemical cues or delivering therapeutics such as cytokines, small molecules, microRNA, growth factors, or stem cells in the damaged tissues. Herein, we present an overview of immunosuppressive modulation by biomaterials in tissue regeneration and highlight the mechanisms of macrophage polarization and Treg cell induction. Overall, we foresee that future biomaterials for regenerative strategies will entail more interactions between biomaterials and the immune cells, and more mechanisms of immunosuppression related to T cell subsets remain to be discovered and applied to develop novel biomaterials for tissue repair and regeneration. STATEMENT OF SIGNIFICANCE: Immunosuppression plays a key role in tissue repair and regeneration, and biomaterials can interact with the immune system through their biological properties and by providing physiochemical cues. Here, we summarize the studies on biomaterials that have been used for immunosuppression to facilitate tissue regeneration. In the first part of this review, we demonstrate the crucial role of macrophage polarization and induction of T regulatory (Treg) cells in immunosuppression. In the second part, distinct approaches used by biomaterials to induce immunosuppression are introduced, which show excellent performance in terms of promoting tissue regeneration.
Collapse
Affiliation(s)
- Peng Shen
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Yanxin Chen
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Shuai Luo
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Zhiyuan Fan
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA 19104, USA
| | - Jilong Wang
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Jiang Chang
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China; State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China.
| | - Junjie Deng
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China.
| |
Collapse
|
31
|
Versey Z, da Cruz Nizer WS, Russell E, Zigic S, DeZeeuw KG, Marek JE, Overhage J, Cassol E. Biofilm-Innate Immune Interface: Contribution to Chronic Wound Formation. Front Immunol 2021; 12:648554. [PMID: 33897696 PMCID: PMC8062706 DOI: 10.3389/fimmu.2021.648554] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/17/2021] [Indexed: 12/19/2022] Open
Abstract
Delayed wound healing can cause significant issues for immobile and ageing individuals as well as those living with co-morbid conditions such as diabetes, cardiovascular disease, and cancer. These delays increase a patient’s risk for infection and, in severe cases, can result in the formation of chronic, non-healing ulcers (e.g., diabetic foot ulcers, surgical site infections, pressure ulcers and venous leg ulcers). Chronic wounds are very difficult and expensive to treat and there is an urgent need to develop more effective therapeutics that restore healing processes. Sustained innate immune activation and inflammation are common features observed across most chronic wound types. However, the factors driving this activation remain incompletely understood. Emerging evidence suggests that the composition and structure of the wound microbiome may play a central role in driving this dysregulated activation but the cellular and molecular mechanisms underlying these processes require further investigation. In this review, we will discuss the current literature on: 1) how bacterial populations and biofilms contribute to chronic wound formation, 2) the role of bacteria and biofilms in driving dysfunctional innate immune responses in chronic wounds, and 3) therapeutics currently available (or underdevelopment) that target bacteria-innate immune interactions to improve healing. We will also discuss potential issues in studying the complexity of immune-biofilm interactions in chronic wounds and explore future areas of investigation for the field.
Collapse
Affiliation(s)
- Zoya Versey
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| | | | - Emily Russell
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| | - Sandra Zigic
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| | - Katrina G DeZeeuw
- Department of Complex Continuing Care, Saint Vincent Hospital, Ottawa, ON, Canada
| | - Jonah E Marek
- Department of Complex Continuing Care, Saint Vincent Hospital, Ottawa, ON, Canada
| | - Joerg Overhage
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| | - Edana Cassol
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada.,Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
32
|
Older but Not Wiser: the Age-Driven Changes in Neutrophil Responses during Pulmonary Infections. Infect Immun 2021; 89:IAI.00653-20. [PMID: 33495271 DOI: 10.1128/iai.00653-20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Elderly individuals are at increased risk of life-threatening pulmonary infections. Neutrophils are a key determinant of the disease course of pathogen-induced pneumonia. Optimal host defense balances initial robust pulmonary neutrophil responses to control pathogen numbers, ultimately followed by the resolution of inflammation to prevent pulmonary damage. Recent evidence suggests that phenotypic and functional heterogeneity in neutrophils impacts host resistance to pulmonary pathogens. Apart from their apparent role in innate immunity, neutrophils also orchestrate subsequent adaptive immune responses during infection. Thus, the outcome of pulmonary infections can be shaped by neutrophils. This review summarizes the age-driven impairment of neutrophil responses and the contribution of these cells to the susceptibility of the elderly to pneumonia. We describe how aging is accompanied by changes in neutrophil recruitment, resolution, and function. We discuss how systemic and local changes alter the neutrophil phenotype in aged hosts. We highlight the gap in knowledge of whether these changes in neutrophils also contribute to the decline in adaptive immunity seen with age. We further detail the factors that drive dysregulated neutrophil responses in the elderly and the pathways that may be targeted to rebalance neutrophil activity and boost host resistance to pulmonary infections.
Collapse
|
33
|
Zhang W, Qu X, Zhu Z, Wang L, Qi Q, Zhou P, Wang X, Li W. Inhibition of miR-139-5p by topical JTXK gel promotes healing of Staphylococcus aureus-infected skin wounds. Cells Dev 2021; 166:203658. [PMID: 33994349 DOI: 10.1016/j.cdev.2021.203658] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/21/2020] [Accepted: 01/04/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND The inflammatory skin wound response is regulated by argonaute 2-bound microRNAs (Ago2-miRNAs) such as miR-139-5p, which inhibit transcription of their target mRNAs. Jiang Tang Xiao Ke (JTXK) is a traditional Chinese medicine that reduces miR-139-5p expression, suggesting that topical application of JTXK may have effects on wound healing. METHODS miR-139-/- mice and wild-type (WT) mice were employed to characterize the in vivo effects of miR-139-5p on sterile wound healing. Neutrophil migration and activation into the wound site were examined by live imaging analysis in lys-EGFP mice and myeloperoxidase/aminophenyl fluorescein assays, respectively. In silico and in vitro studies in differentiated HL60 cells were performed to identify miR-139-5p's downstream mediator(s). miR-139-/- neutrophil transplantation (with or without Eif4g2-knockdown rescue) or a topical JTXK gel preparation (with or without miR-139-5p mimic rescue) were employed to characterize the in vivo effects of miR-139-5p and JTXK, respectively, on Staphylococcus aureus (S. aureus)-infected wound healing. RESULTS miR-139-/- mice display impaired sterile wound healing but improved S. aureus-infected wound healing. Eif4g2, a protein that supports neutrophil proliferation and differentiation, was identified as a key downstream mediator of miR-139-5p. miR-139-/- mice show elevated neutrophilic activation and Eif4g2 upregulation. miR-139-/- neutrophils enhanced S. aureus-infected wound healing in an Eif4g2-dependent manner. Moreover, topical JTXK gel therapy also enhanced S. aureus-infected wound healing in a miR-139-5p-dependent manner. CONCLUSIONS miR-139-5p negatively regulates the neutrophilic response during S. aureus-infected wound healing, suggesting that JTXK or other miR-139-5p suppressants may be effective for treating infected skin wounds.
Collapse
Affiliation(s)
- Weitao Zhang
- Department of Dermatology, Affiliated Hospital of Shaanxi University of traditional Chinese Medicine, Xianyang, China.
| | - Xu Qu
- Department of Dermatology, Affiliated Hospital of Shaanxi University of traditional Chinese Medicine, Xianyang, China
| | - Zhang Zhu
- Department of Dermatology, Affiliated Hospital of Shaanxi University of traditional Chinese Medicine, Xianyang, China
| | - Liwen Wang
- Department of Dermatology, Affiliated Hospital of Shaanxi University of traditional Chinese Medicine, Xianyang, China
| | - Qian Qi
- Shaanxi University of Traditional Chinese Medicine, Xianyang, China
| | - Pengjun Zhou
- Department of Dermatology, Affiliated Hospital of Shaanxi University of traditional Chinese Medicine, Xianyang, China
| | - Xiaoli Wang
- Department of Dermatology, Affiliated Hospital of Shaanxi University of traditional Chinese Medicine, Xianyang, China
| | - Wenna Li
- Department of Dermatology, Affiliated Hospital of Shaanxi University of traditional Chinese Medicine, Xianyang, China
| |
Collapse
|
34
|
Genome-Scale Transcript Analyses of Human Neutrophils. Methods Mol Biol 2021. [PMID: 31728999 DOI: 10.1007/978-1-0716-0154-9_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Transcriptome analyses of unicellular and multicellular organisms have changed fundamental understanding of biological and pathological processes across multiple scientific disciplines. Over the past 15 years, studies of polymorphonuclear leukocyte (PMN or neutrophil) gene expression on a global scale have provided new insight into the molecular processes that promote resolution of infections in humans. Herein we present methods to analyze gene expression in human neutrophils using Affymetrix oligonucleotide microarrays and next-generation sequencing. Notably, the procedures utilize commercially available reagents and materials and thus represent a standardized approach for evaluating PMN transcript levels.
Collapse
|
35
|
Liu Y, Kaplan MJ. Neutrophils in the Pathogenesis of Rheumatic Diseases: Fueling the Fire. Clin Rev Allergy Immunol 2020; 60:1-16. [DOI: 10.1007/s12016-020-08816-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 12/11/2022]
|
36
|
Cell type specific gene expression profiling reveals a role for complement component C3 in neutrophil responses to tissue damage. Sci Rep 2020; 10:15716. [PMID: 32973200 PMCID: PMC7518243 DOI: 10.1038/s41598-020-72750-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/27/2020] [Indexed: 01/09/2023] Open
Abstract
Tissue damage induces rapid recruitment of leukocytes and changes in the transcriptional landscape that influence wound healing. However, the cell-type specific transcriptional changes that influence leukocyte function and tissue repair have not been well characterized. Here, we employed translating ribosome affinity purification (TRAP) and RNA sequencing, TRAP-seq, in larval zebrafish to identify genes differentially expressed in neutrophils, macrophages, and epithelial cells in response to wounding. We identified the complement pathway and c3a.1, homologous to the C3 component of human complement, as significantly increased in neutrophils in response to wounds. c3a.1−/− zebrafish larvae have impaired neutrophil directed migration to tail wounds with an initial lag in recruitment early after wounding. Moreover, c3a.1−/− zebrafish larvae have impaired recruitment to localized bacterial infections and reduced survival that is, at least in part, neutrophil mediated. Together, our findings support the power of TRAP-seq to identify cell type specific changes in gene expression that influence neutrophil behavior in response to tissue damage.
Collapse
|
37
|
Oba J, Okabe M, Yoshida T, Soko C, Fathy M, Amano K, Kobashi D, Wakasugi M, Okudera H. Hyperdry human amniotic membrane application as a wound dressing for a full-thickness skin excision after a third-degree burn injury. BURNS & TRAUMA 2020; 8:tkaa014. [PMID: 32733973 PMCID: PMC7382972 DOI: 10.1093/burnst/tkaa014] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/08/2020] [Indexed: 12/18/2022]
Abstract
Background Severe burn injuries create large skin defects that render the host susceptible to bacterial infections. Burn wound infection often causes systemic sepsis and severe septicemia, resulting in an increase in the mortality of patients with severe burn injuries. Therefore, appropriate wound care is important to prevent infection and improve patient outcomes. However, it is difficult to heal a third-degree burn injury. The aim of this study was to investigate whether hyperdry human amniotic membrane (HD-AM) could promote early granulation tissue formation after full-thickness skin excision in third-degree burn injury sites in mice. Methods After the development of HD-AM and creation of a third-degree burn injury model, the HD-AM was either placed or not placed on the wound area in the HD-AM group or HD-AM group, respectively. The groups were prepared for evaluation on postoperative days 1, 4 and 7. Azan staining was used for granulation tissue evaluation, and estimation of CD163, transforming growth factor beta-1 (TGF-β1), vascular endothelial growth factor (VEGF), CD31, alpha-smooth muscle actin (α-SMA) and Iba1 expression was performed by immunohistochemical staining. Quantitative reverse-transcription polymerase chain reaction (PCR) was used to investigate gene expression of growth factors, cell migration chemokines and angiogenic and inflammatory markers. Results The HD-AM group showed significant early and qualitatively good growth of granulation tissue on the full-thickness skin excision site. HD-AM promoted early-phase inflammatory cell infiltration, fibroblast migration and angiogenesis in the granulation tissue. Additionally, the early infiltration of cells of the immune system was observed. Conclusions HD-AM may be useful as a new wound dressing material for full-thickness skin excision sites after third-degree burn injuries, and may be a new therapeutic technique for improving the survival rate of patients with severe burn injuries.
Collapse
Affiliation(s)
- Jiro Oba
- Department of Emergency and Disaster Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Motonori Okabe
- Department of Regenerative Medicine, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Toshiko Yoshida
- Department of Regenerative Medicine, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Chika Soko
- Department of Regenerative Medicine, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Moustafa Fathy
- Department of Regenerative Medicine, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan.,Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Koji Amano
- Department of Emergency Surgery, Sakai City Medical Center, Sakai, Osaka 594-8304, Japan
| | - Daisuke Kobashi
- Department of Emergency and Disaster Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Masahiro Wakasugi
- Department of Emergency and Disaster Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Hiroshi Okudera
- Department of Emergency and Disaster Medicine, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
38
|
Fetz AE, Radic MZ, Bowlin GL. Neutrophils in Biomaterial-Guided Tissue Regeneration: Matrix Reprogramming for Angiogenesis. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:95-106. [PMID: 32299302 DOI: 10.1089/ten.teb.2020.0028] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Biomaterial-guided in situ tissue regeneration uses biomaterials to stimulate and guide the body's endogenous, regenerative processes to drive functional tissue repair and regeneration. To be successful, cell migration into the biomaterials is essential, which requires angiogenesis to maintain cell viability. Neutrophils, the first cells responding to an implanted biomaterial, are now known to play an integral part in angiogenesis in multiple tissues and exhibit considerable potential for driving angiogenesis in the context of tissue regeneration. In terms of biomaterial-guided in situ tissue regeneration, harnessing the proangiogenic potential of the neutrophil through its robust secretion of matrix metalloproteinase 9 (MMP-9) may provide a mechanism to improve biomaterial performance by initiating matrix reprogramming. This review will discuss neutrophils as matrix reprogrammers and what is currently known about their ability to create a microenvironment that is more conducive for angiogenesis and tissue regeneration through the secretion of MMP-9. It will first review a set of ground-breaking studies in tumor biology and then present an overview of what is currently known about neutrophils and MMP-9 in biomaterial vascularization. Finally, it will conclude with potential strategies and considerations to engage neutrophils in biomaterial-guided angiogenesis and in situ tissue regeneration. Impact statement This review draws attention to a highly neglected topic in tissue engineering, the role of neutrophils in biomaterial-guided tissue regeneration and angiogenesis. Moreover, it highlights their abundant secretion of matrix metalloproteinase 9 (MMP-9) for matrix reprogramming, a topic with great potential yet to be vetted in the literature. It presents strategies and considerations for designing the next generation of immunomodulatory biomaterials. While there is literature discussing the overall role of neutrophils in angiogenesis, there are a limited number of review articles focused on this highly relevant topic in the context of biomaterial integration and tissue regeneration, making this a necessary and impactful article.
Collapse
Affiliation(s)
- Allison E Fetz
- Department of Biomedical Engineering, University of Memphis, Memphis, Tennessee, USA
| | - Marko Z Radic
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Gary L Bowlin
- Department of Biomedical Engineering, University of Memphis, Memphis, Tennessee, USA
| |
Collapse
|
39
|
Joshi N, Pohlmeier L, Ben-Yehuda Greenwald M, Haertel E, Hiebert P, Kopf M, Werner S. Comprehensive characterization of myeloid cells during wound healing in healthy and healing-impaired diabetic mice. Eur J Immunol 2020; 50:1335-1349. [PMID: 32306381 PMCID: PMC7496577 DOI: 10.1002/eji.201948438] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/18/2020] [Accepted: 04/16/2020] [Indexed: 01/13/2023]
Abstract
Wound healing involves the concerted action of various lymphoid and in particular myeloid cell populations. To characterize and quantitate different types of myeloid cells and to obtain information on their kinetics during wound healing, we performed multiparametric flow cytometry analysis. In healthy mice, neutrophil numbers increased early after injury and returned to near basal levels after completion of healing. Macrophages, monocyte‐derived dendritic cells (DCs), and eosinophils were abundant throughout the healing phase, in particular in early wounds, and Langerhans cells increased after wounding and remained elevated after epithelial closure. Major differences in healing‐impaired diabetic mice were a much higher percentage of immune cells in late wounds, mainly as a result of neutrophil, macrophage, and monocyte persistence; reduced numbers and percentages of macrophages and monocyte‐derived DCs in early wounds; and of Langerhans cells, conventional DCs, and eosinophils throughout the healing process. Finally, unbiased cluster analysis (PhenoGraph) identified a large number of different clusters of myeloid cells in skin wounds. These results provide insight into myeloid cell diversity and dynamics during wound repair and highlight the abnormal inflammatory response associated with impaired healing.
Collapse
Affiliation(s)
- Natasha Joshi
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Lea Pohlmeier
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | | | - Eric Haertel
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Paul Hiebert
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
40
|
Overview of Basic Immunology and Clinical Application. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1244:1-36. [PMID: 32301008 DOI: 10.1007/978-3-030-41008-7_1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tumor exists as a complex network of structures with an ability to evolve and evade the host immune surveillance mechanism. The immune milieu which includes macrophages, dendritic cells, natural killer cells, neutrophils, mast cells, B cells, and T cells are found in the core, the invasive margin, or the adjacent stromal or lymphoid component of the tumor. The immune infiltrate is heterogeneous and varies within a patient and between patients of the same tumor histology. The location, density, functionality, and cross-talk between the immune cells in the tumor microenvironment influence the nature of immune response, prognosis, and treatment outcomes in cancer patients. Therefore, an understanding of the characteristics of the immune cells and their role in tumor immune surveillance is of paramount importance to identify immune targets and to develop novel immune therapeutics in the war against cancer. In this chapter, we provide an overview of the individual components of the human immune system and the translational relevance of predictive biomarkers.
Collapse
|
41
|
Abstract
Neutrophils, also known as polymorphonuclear neutrophils (PMNs), have long been considered as the short-lived, nonspecific white cells that form pus-and also happen to kill invading microbes. Indeed, neutrophils were often neglected (and largely not considered) as immune cells. This historic view of neutrophils has changed considerably over the past several decades, and we now know that in addition to playing the predominant role in the clearance of bacteria and fungi, they have a major role in shaping the host response to infection and immune system homeostasis. The change in our view of the role of neutrophils in the immune system has been due in large part to the study of these cells in vitro. Such work has been made possible by new and/or improved methods and approaches used to investigate neutrophils. These methods are the focus of this volume.
Collapse
Affiliation(s)
- Harry L Malech
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Frank R DeLeo
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Mark T Quinn
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA.
| |
Collapse
|
42
|
The pathogenesis of systemic lupus erythematosus: Harnessing big data to understand the molecular basis of lupus. J Autoimmun 2019; 110:102359. [PMID: 31806421 DOI: 10.1016/j.jaut.2019.102359] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/04/2019] [Indexed: 12/22/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic, systemic autoimmune disease that causes damage to multiple organ systems. Despite decades of research and available murine models that capture some aspects of the human disease, new treatments for SLE lag behind other autoimmune diseases such as Rheumatoid Arthritis and Crohn's disease. Big data genomic assays have transformed our understanding of SLE by providing important insights into the molecular heterogeneity of this multigenic disease. Gene wide association studies have demonstrated more than 100 risk loci, supporting a model of multiple genetic hits increasing SLE risk in a non-linear fashion, and providing evidence of ancestral diversity in susceptibility loci. Epigenetic studies to determine the role of methylation, acetylation and non-coding RNAs have provided new understanding of the modulation of gene expression in SLE patients and identified new drug targets and biomarkers for SLE. Gene expression profiling has led to a greater understanding of the role of myeloid cells in the pathogenesis of SLE, confirmed roles for T and B cells in SLE, promoted clinical trials based on the prominent interferon signature found in SLE patients, and identified candidate biomarkers and cellular signatures to further drug development and drug repurposing. Gene expression studies are advancing our understanding of the underlying molecular heterogeneity in SLE and providing hope that patient stratification will expedite new therapies based on personal molecular signatures. Although big data analyses present unique interpretation challenges, both computationally and biologically, advances in machine learning applications may facilitate the ability to predict changes in SLE disease activity and optimize therapeutic strategies.
Collapse
|
43
|
Brazil JC, Quiros M, Nusrat A, Parkos CA. Innate immune cell-epithelial crosstalk during wound repair. J Clin Invest 2019; 129:2983-2993. [PMID: 31329162 PMCID: PMC6668695 DOI: 10.1172/jci124618] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Skin and intestinal epithelial barriers play a pivotal role in protecting underlying tissues from harsh external environments. The protective role of these epithelia is, in part, dependent on a remarkable capacity to restore barrier function and tissue homeostasis after injury. In response to damage, epithelial wounds repair by a series of events that integrate epithelial responses with those of resident and infiltrating immune cells including neutrophils and monocytes/macrophages. Compromise of this complex interplay predisposes to development of chronic nonhealing wounds, contributing to morbidity and mortality of many diseases. Improved understanding of crosstalk between epithelial and immune cells during wound repair is necessary for development of better pro-resolving strategies to treat debilitating complications of disorders ranging from inflammatory bowel disease to diabetes. In this Review we focus on epithelial and innate immune cell interactions that mediate wound healing and restoration of tissue homeostasis in the skin and intestine.
Collapse
|
44
|
Rungelrath V, Kobayashi SD, DeLeo FR. Neutrophils in innate immunity and systems biology-level approaches. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2019; 12:e1458. [PMID: 31218817 DOI: 10.1002/wsbm.1458] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022]
Abstract
The innate immune system is the first line of host defense against invading microorganisms. Polymorphonuclear leukocytes (PMNs or neutrophils) are the most abundant leukocyte in humans and essential to the innate immune response against invading pathogens. Compared to the acquired immune response, which requires time to develop and is dependent on previous interaction with specific microbes, the ability of neutrophils to kill microorganisms is immediate, nonspecific, and not dependent on previous exposure to microorganisms. Historically, studies of PMN-pathogen interaction focused on the events leading to killing of microorganisms, such as recruitment/chemotaxis, transmigration, phagocytosis, and activation, whereas postphagocytosis sequelae were infrequently considered. In addition, it was widely accepted that human neutrophils possessed limited capacity for new gene transcription and thus, relatively little biosynthetic capacity. This notion has changed dramatically within the past 20 years. Further, there is now more effort directed to understand the events occurring in PMNs after killing of microbes. Herein, we give an updated review of the systems biology-level approaches that have been used to gain an enhanced view of the role of neutrophils during host-pathogen interaction and neutrophil-mediated diseases. We anticipate that these and future systems-level studies will continue to provide information important for understanding, treatment, and control of diseases caused by pathogenic microorganisms. This article is categorized under: Physiology > Organismal Responses to Environment Physiology > Mammalian Physiology in Health and Disease Biological Mechanisms > Cell Fates.
Collapse
Affiliation(s)
- Viktoria Rungelrath
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Scott D Kobayashi
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Frank R DeLeo
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| |
Collapse
|
45
|
Peiseler M, Kubes P. More friend than foe: the emerging role of neutrophils in tissue repair. J Clin Invest 2019; 129:2629-2639. [PMID: 31205028 DOI: 10.1172/jci124616] [Citation(s) in RCA: 222] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Neutrophils are the most abundant immune cells in humans and serve as first responders to a myriad of host perturbations. Equipped with a plethora of antimicrobial molecules, neutrophils invade sites of inflammation to eradicate pathogens and clear debris. Traditionally, neutrophils were thought to cause collateral tissue damage before dying at the site. However, the presence of neutrophil infiltration into sterile injuries (in the absence of infections) suggests additional roles for these cells. Now, the view of neutrophils as indiscriminate killers seems to be changing as evolving evidence suggests that neutrophils actively orchestrate resolution of inflammation and contribute to tissue repair. Novel concepts include the idea that neutrophils are key to revascularization and subsequently reverse-transmigrate back to the vasculature, actively leaving sites of tissue damage to re-home to functional niches in the lung and bone marrow. This Review scrutinizes the role of neutrophils in tissue damage and repair, discussing recent findings and raising unresolved questions around this intriguing immune cell.
Collapse
Affiliation(s)
- Moritz Peiseler
- Department of Pharmacology and Physiology.,Snyder Institute for Chronic Diseases, and
| | - Paul Kubes
- Department of Pharmacology and Physiology.,Snyder Institute for Chronic Diseases, and.,Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
46
|
SIRT3 Regulates Macrophage-Mediated Inflammation in Diabetic Wound Repair. J Invest Dermatol 2019; 139:2528-2537.e2. [PMID: 31207226 DOI: 10.1016/j.jid.2019.05.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 05/22/2019] [Accepted: 05/28/2019] [Indexed: 12/26/2022]
Abstract
Control of inflammation is critical for the treatment of nonhealing wounds, but a delicate balance exists between early inflammation that is essential for normal tissue repair and the pathologic inflammation that can occur later in the repair process. This necessitates the development of novel therapies that can target inflammation at the appropriate time during repair. Here, we found that SIRT3 is essential for normal healing and regulates inflammation in wound macrophages after injury. Under prediabetic conditions, SIRT3 was decreased in wound macrophages and resulted in dysregulated inflammation. In addition, we found that FABP4 regulates SIRT3 in human blood monocytes, and inhibition of FABP4 in wound macrophages decreases inflammatory cytokine expression, making FABP4 a viable target for the regulation of excess inflammation and wound repair in diabetes. Using a series of ex vivo and in vivo studies with genetically engineered mouse models and diabetic human monocytes, we showed that FABP4 expression is epigenetically upregulated in diabetic wound macrophages and, in turn, diminishes SIRT3 expression, thereby promoting inflammation. These findings have significant implications for controlling inflammation and promoting tissue repair in diabetic wounds.
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW This review describes the contribution of coagulation factor XII (FXII) in sterile inflammation and wound healing, focusing on recently identified roles for zymogen FXII in neutrophil functions. RECENT FINDINGS Recent studies have identified an important role for FXII in neutrophil trafficking. In particular, following neutrophil activation, autocrine FXII signals through the urokinase plasminogen activator receptor (uPAR) on the neutrophil surface to upregulate neutrophil functions. The sum of these activities leads to neutrophil adhesion, chemotaxis, and neutrophil extracellular (NET) formation. Downregulating FXII-mediated signaling in neutrophils is associated with improved wound healing. SUMMARY These recent findings show the sophisticated role of FXII in vivo and create new opportunities for research on the treatment of chronic inflammatory diseases.
Collapse
|
48
|
The Dynamics of the Skin's Immune System. Int J Mol Sci 2019; 20:ijms20081811. [PMID: 31013709 PMCID: PMC6515324 DOI: 10.3390/ijms20081811] [Citation(s) in RCA: 343] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 12/12/2022] Open
Abstract
The skin is a complex organ that has devised numerous strategies, such as physical, chemical, and microbiological barriers, to protect the host from external insults. In addition, the skin contains an intricate network of immune cells resident to the tissue, crucial for host defense as well as tissue homeostasis. In the event of an insult, the skin-resident immune cells are crucial not only for prevention of infection but also for tissue reconstruction. Deregulation of immune responses often leads to impaired healing and poor tissue restoration and function. In this review, we will discuss the defensive components of the skin and focus on the function of skin-resident immune cells in homeostasis and their role in wound healing.
Collapse
|
49
|
Fischer J, Walter C, Tönges A, Aleth H, Jordão MJC, Leddin M, Gröning V, Erdmann T, Lenz G, Roth J, Vogl T, Prinz M, Dugas M, Jacobsen ID, Rosenbauer F. Safeguard function of PU.1 shapes the inflammatory epigenome of neutrophils. Nat Immunol 2019; 20:546-558. [DOI: 10.1038/s41590-019-0343-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 02/08/2019] [Indexed: 12/16/2022]
|
50
|
den Dekker A, Davis FM, Kunkel SL, Gallagher KA. Targeting epigenetic mechanisms in diabetic wound healing. Transl Res 2019; 204:39-50. [PMID: 30392877 PMCID: PMC6331222 DOI: 10.1016/j.trsl.2018.10.001] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 10/02/2018] [Accepted: 10/02/2018] [Indexed: 02/07/2023]
Abstract
Impaired wound healing is a major secondary complication of type 2 diabetes that often results in limb loss and disability. Normal tissue repair progresses through discrete phases including hemostasis, inflammation, proliferation, and remodeling. In diabetes, normal progression through these phases is impaired resulting in a sustained inflammatory state and dysfunctional epithelialization in the wound. Due to their plasticity, macrophages play a critical role in the transition from the inflammation phase to the proliferation phase. Diabetes disrupts macrophage function by impairing monocyte recruitment to the wound, reducing phagocytosis, and prohibiting the transition of inflammatory macrophages to an anti-inflammatory state. Diabetes also impedes keratinocyte and fibroblast function during the later phases resulting in impaired epithelialization of the wound. Several recent studies suggest that altered epigenetic regulation of both immune and structural cells in wounds may influence cell phenotypes and healing, particularly in pathologic states, such as diabetes. Specifically, it has been shown that macrophage plasticity during wound repair is partly regulated epigenetically and that diabetes alters this epigenetic regulation and contributes to a sustained inflammatory state. Epigenetic regulation is also known to regulate keratinocyte and fibroblast function during wound repair. In this review, we provide an introduction to the epigenetic mechanisms that regulate tissue repair and highlight recent findings that demonstrate, how epigenetic events are altered during the course of diabetic wound healing.
Collapse
Affiliation(s)
- Aaron den Dekker
- Department of Surgery, University of Michigan, Ann Arbor, Michgan
| | - Frank M Davis
- Department of Surgery, University of Michigan, Ann Arbor, Michgan
| | - Steve L Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|