1
|
Zhang J, Shimozaki K, Hattori S, Pastukh V, Maloney D, Hogan MV, Wang JHC. Metformin lotion promotes scarless skin tissue formation through AMPK activation, TGF-β1 inhibition, and reduced myofibroblast numbers. PLoS One 2024; 19:e0311147. [PMID: 39331598 PMCID: PMC11433050 DOI: 10.1371/journal.pone.0311147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/10/2024] [Indexed: 09/29/2024] Open
Abstract
Scar tissue formation following skin wound healing is a challenging public health problem. Skin regeneration and preventing the formation of scar tissue by currently available commercial products are largely ineffective. This study aimed to test the efficacy of a novel topical metformin lotion (ML) in inhibiting scar tissue formation during skin wound healing in rats and to determine the mechanisms of action involved. A 6% ML was prepared in our laboratory. A skin wound healing model in rats was used. The wounded rats were divided into two groups and treated daily for 10 days as follows: Group 1 received a daily application of 50 mg of control lotion, or 0% ML (totaling 100 mg of lotion per rat), and Group 2 received a daily application of 50 mg of 6% ML (totaling 100 mg of 6% ML per rat). Blood samples from the heart of each rat were analyzed for inflammatory markers, HMGB1 and IL-1β, using ELISA, and immunological and histological analyses were performed on skin tissue sections. ML decreased levels of inflammatory markers HMGB1 and IL-1β in the serum of rats and inhibited the release of HMGB1 from cell nuclei into the skin tissue matrix. Additionally, ML demonstrated anti-fibrotic properties by enhancing AMPK activity, decreasing the expression of TGF-β1, reducing the number of myofibroblasts, decreasing the production of collagen III, and increasing the expression of collagen I. ML promotes the regeneration of high-quality skin during wound healing by reducing scar tissue formation. This effect is mediated through the activation of AMPK, inhibition of TGF-β1, and a decrease in the number of myofibroblasts.
Collapse
Affiliation(s)
- Jianying Zhang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kengo Shimozaki
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Soichi Hattori
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Vasyl Pastukh
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Derek Maloney
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - MaCalus V. Hogan
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - James H-C. Wang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
2
|
Li S, Tao G. Perish in the Attempt: Regulated Cell Death in Regenerative and Nonregenerative Tissue. Antioxid Redox Signal 2023; 39:1053-1069. [PMID: 37218435 PMCID: PMC10715443 DOI: 10.1089/ars.2022.0166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/24/2023]
Abstract
Significance: A cell plays its roles throughout its life span, even during its demise. Regulated cell death (RCD) is one of the key topics in modern biomedical studies. It is considered the main approach for removing stressed and/or damaged cells. Research during the past two decades revealed more roles of RCD, such as coordinating tissue development and driving compensatory proliferation during tissue repair. Recent Advances: Compensatory proliferation, initially identified in primitive organisms during the regeneration of lost tissue, is an evolutionarily conserved process that also functions in mammals. Among various types of RCD, apoptosis is considered the top candidate to induce compensatory proliferation in damaged tissue. Critical Issues: The roles of apoptosis in the recovery of nonregenerative tissue are still vague. The roles of other types of RCD, such as necroptosis and ferroptosis, have not been well characterized in the context of tissue regeneration. Future Directions: In this review article, we attempt to summarize the recent insights on the role of RCD in tissue repair. We focus on apoptosis, with expansion to ferroptosis and necroptosis, in primitive organisms with significant regenerative capacity as well as common mammalian research models. After gathering hints from regenerative tissue, in the second half of the review, we take a notoriously nonregenerative tissue, the myocardium, as an example to discuss the role of RCD in terminally differentiated quiescent cells. Antioxid. Redox Signal. 39, 1053-1069.
Collapse
Affiliation(s)
- Shuang Li
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
3
|
Enhanced Wound Healing Potential of Spirulina platensis Nanophytosomes: Metabolomic Profiling, Molecular Networking, and Modulation of HMGB-1 in an Excisional Wound Rat Model. Mar Drugs 2023; 21:md21030149. [PMID: 36976198 PMCID: PMC10051127 DOI: 10.3390/md21030149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
Excisional wounds are considered one of the most common physical injuries. This study aims to test the effect of a nanophytosomal formulation loaded with a dried hydroalcoholic extract of S. platensis on promoting excisional wound healing. The Spirulina platensis nanophytosomal formulation (SPNP) containing 100 mg PC and 50 mg CH exhibited optimum physicochemical characteristics regarding particle size (598.40 ± 9.68 nm), zeta potential (−19.8 ± 0.49 mV), entrapment efficiency (62.76 ± 1.75%), and Q6h (74.00 ± 1.90%). It was selected to prepare an HPMC gel (SPNP-gel). Through metabolomic profiling of the algal extract, thirteen compounds were identified. Molecular docking of the identified compounds on the active site of the HMGB-1 protein revealed that 12,13-DiHome had the highest docking score of −7.130 kcal/mol. SPNP-gel showed higher wound closure potential and enhanced histopathological alterations as compared to standard (MEBO® ointment) and S. platensis gel in wounded Sprague-Dawley rats. Collectively, NPS promoted the wound healing process by enhancing the autophagy process (LC3B/Beclin-1) and the NRF-2/HO-1antioxidant pathway and halting the inflammatory (TNF-, NF-κB, TlR-4 and VEGF), apoptotic processes (AIF, Caspase-3), and the downregulation of HGMB-1 protein expression. The present study’s findings suggest that the topical application of SPNP-gel possesses a potential therapeutic effect in excisional wound healing, chiefly by downregulating HGMB-1 protein expression.
Collapse
|
4
|
Shershakova NN, Andreev SM, Tomchuk AA, Makarova EA, Nikonova AA, Turetskiy EA, Petukhova OA, Kamyshnikov OY, Ivankov OI, Kyzyma OA, Tomchuk OV, Avdeev MV, Dvornikov AS, Kudlay DA, Khaitov MR. Wound healing activity of aqueous dispersion of fullerene C 60 produced by "green technology". NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 47:102619. [PMID: 36272619 DOI: 10.1016/j.nano.2022.102619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 08/13/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
In addition to exhibited antioxidant and anti-inflammatory activity, fullerene C60 is a promising wound healing agent. An important stage in the production of fullerene-based ointments is the stability of the aqueous fullerene dispersion (AFD) with minimum size of colloidal fullerene aggregates and sufficiently high concentration. To achieve these parameters tangential flow filtration of fullerene C60 was used ("green technology"). As estimated by small-angle neutron scattering and dynamic light scattering purified AFDs with narrow-size distribution nanoclusters have a size of 6 nm and are assembled into agglomerates which reach a size of 150 nm. The ability of the AFD to exhibit regenerative activity was studied using the animal wound model. This study shows for the first time that the fullerene-based composition stimulates the healing of wounds of various origins. We assume that the mechanism of the AFD wound-healing activity is associated with the aryl hydrocarbon receptor and macrophages activity.
Collapse
Affiliation(s)
- N N Shershakova
- NRC Institute of Immunology FMBA of Russia, Kashirskoe shosse, 24, Moscow 115522, Russian Federation.
| | - S M Andreev
- NRC Institute of Immunology FMBA of Russia, Kashirskoe shosse, 24, Moscow 115522, Russian Federation
| | - A A Tomchuk
- International intergovernmental organization Joint Institute for Nuclear Research, st. Joliot-Curie, 6, Dubna, Moscow Region 141980, Russian Federation
| | - E A Makarova
- NRC Institute of Immunology FMBA of Russia, Kashirskoe shosse, 24, Moscow 115522, Russian Federation
| | - A A Nikonova
- NRC Institute of Immunology FMBA of Russia, Kashirskoe shosse, 24, Moscow 115522, Russian Federation
| | - E A Turetskiy
- NRC Institute of Immunology FMBA of Russia, Kashirskoe shosse, 24, Moscow 115522, Russian Federation
| | - O A Petukhova
- NRC Institute of Immunology FMBA of Russia, Kashirskoe shosse, 24, Moscow 115522, Russian Federation
| | - O Y Kamyshnikov
- NRC Institute of Immunology FMBA of Russia, Kashirskoe shosse, 24, Moscow 115522, Russian Federation
| | - O I Ivankov
- International intergovernmental organization Joint Institute for Nuclear Research, st. Joliot-Curie, 6, Dubna, Moscow Region 141980, Russian Federation; Moscow Institute of Physics and Technology, 9 Institutskiy per., Dolgoprudny, Moscow Region 141701, Russian Federation
| | - O A Kyzyma
- Taras Shevchenko National University of Kyiv, 64/13, Volodymyrska Street, Kyiv 0160, Ukraine
| | - O V Tomchuk
- International intergovernmental organization Joint Institute for Nuclear Research, st. Joliot-Curie, 6, Dubna, Moscow Region 141980, Russian Federation; Taras Shevchenko National University of Kyiv, 64/13, Volodymyrska Street, Kyiv 0160, Ukraine
| | - M V Avdeev
- International intergovernmental organization Joint Institute for Nuclear Research, st. Joliot-Curie, 6, Dubna, Moscow Region 141980, Russian Federation
| | - A S Dvornikov
- Pirogov Russian National Research Medical University, 1 Ostrovityanov St., Moscow 119997, Russian Federation
| | - D A Kudlay
- NRC Institute of Immunology FMBA of Russia, Kashirskoe shosse, 24, Moscow 115522, Russian Federation; I.M. Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya str., Moscow 119991, Russian Federation
| | - M R Khaitov
- NRC Institute of Immunology FMBA of Russia, Kashirskoe shosse, 24, Moscow 115522, Russian Federation; Pirogov Russian National Research Medical University, 1 Ostrovityanov St., Moscow 119997, Russian Federation
| |
Collapse
|
5
|
Cell death in skin function, inflammation, and disease. Biochem J 2022; 479:1621-1651. [PMID: 35929827 PMCID: PMC9444075 DOI: 10.1042/bcj20210606] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022]
Abstract
Cell death is an essential process that plays a vital role in restoring and maintaining skin homeostasis. It supports recovery from acute injury and infection and regulates barrier function and immunity. Cell death can also provoke inflammatory responses. Loss of cell membrane integrity with lytic forms of cell death can incite inflammation due to the uncontrolled release of cell contents. Excessive or poorly regulated cell death is increasingly recognised as contributing to cutaneous inflammation. Therefore, drugs that inhibit cell death could be used therapeutically to treat certain inflammatory skin diseases. Programmes to develop such inhibitors are already underway. In this review, we outline the mechanisms of skin-associated cell death programmes; apoptosis, necroptosis, pyroptosis, NETosis, and the epidermal terminal differentiation programme, cornification. We discuss the evidence for their role in skin inflammation and disease and discuss therapeutic opportunities for targeting the cell death machinery.
Collapse
|
6
|
Zhang K, Yang C, Cheng C, Shi C, Sun M, Hu H, Shi T, Chen X, He X, Zheng X, Li M, Shao D. Bioactive Injectable Hydrogel Dressings for Bacteria-Infected Diabetic Wound Healing: A "Pull-Push" Approach. ACS APPLIED MATERIALS & INTERFACES 2022; 14:26404-26417. [PMID: 35649246 DOI: 10.1021/acsami.2c04300] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Chronic diabetic wound healing remains a challenge due to the existence of excessive danger molecules and bacteria in the inflammatory microenvironment. There is an urgent need for advanced wound dressings that target both inflammation and infection. Here, a bioactive hydrogel without loading any anti-inflammatory ingredients is rationally designed to achieve a "Pull-Push" approach for efficient and safe bacteria-infected diabetic wound healing by integrating danger molecule scavenging (Pull) with antibiotic delivery (Push) in the inflammatory microenvironment. The cationic hydrogel, termed the OCMC-Tob/PEI hydrogel, is fabricated by the conjugation of polyethylenimine (PEI) and tobramycin (Tob) on an oxidized carboxymethyl cellulose (OCMC) backbone via the Schiff base reaction with injectable, self-healing, and biocompatible properties. The OCMC-Tob/PEI hydrogel not only displays the remarkable capability of capturing multiple negatively charged danger molecules (e.g., cell-free DNA, lipopolysaccharides, and tumor necrosis factor-α) to ameliorate anti-inflammation effects but also achieves controllable long-term antibacterial activity by the pH-sensitive release of Tob. Consequently, this multifunctional hydrogel greatly expedites the wound closure rate with combined anti-inflammation and anti-infection effects on Pseudomonas aeruginosa-infected diabetic wounds. Our work provides a highly versatile treatment approach for chronic diabetic wounds and a promising dressing for regenerative medicine.
Collapse
Affiliation(s)
- Kunbao Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong 510006, China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, China
| | - Chao Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong 510006, China
| | - Chuanxu Cheng
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China
| | - Chengxin Shi
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310000, China
| | - Madi Sun
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong 510006, China
| | - Hanze Hu
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Tongfei Shi
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong 510006, China
| | - Xuenian Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong 510006, China
| | - Xuan He
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong 510006, China
| | - Xiao Zheng
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong 510006, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Dan Shao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 511442, China
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong 510006, China
| |
Collapse
|
7
|
Abd-Elhakim YM, Omran BHF, Ezzeldein SA, Ahmed AI, El-Sharkawy NI, Mohamed AAR. Time-dependent expression of high-mobility group box-1 and toll-like receptors proteins as potential determinants of skin wound age in rats: Forensic implication. Int J Legal Med 2022; 136:1781-1789. [PMID: 35132471 PMCID: PMC9576669 DOI: 10.1007/s00414-022-02788-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/21/2022] [Indexed: 12/03/2022]
Abstract
The skin wound age determination in living subjects is an imperative task for forensic experts. In this study, we investigated the time-dependent expression of high-mobility group box-1 (HMGB1) and toll-like receptors 2 and 4 (TLR2 and 4) in rat skin wounds using real-time PCR and seek their forensic potentials during the skin wound repair process. In addition, the levels of serum pro-inflammatory cytokines (tumor necrosis factor-alpha (TNF-α) and interleukin 6 (IL-6)), as well as nitric oxide (NO) production, were measured. The wound tissue and serum samples were collected after 30 min, 2 h, 6 h, 12 h, 1 day, 3 days, 5 days, and 7 days after incision. As a control (zero time), skin specimens and blood samples were collected without incision. The results reveal that the HMGB1, TLR2, and TLR4 expression levels were increased in a time-dependent manner until the first day where the peak level was achieved for the three tested genes compared with the zero time. On the 7th day, the statistical significance was lost for TLR2 and TLR4 but persisted for HMGB1. The serum TNF-α, IL6, and NO levels peaked within 30 min and 1st and 3rd day after injury, respectively. On the 7th day after incision, no significant differences exist in the TNF-α serum level compared to the control group, but the statistical significance persisted for IL6 and NO. It was apparent that the analyzed genes in the wound tissues showed higher R2 values rather than the serum biochemical indicators. Of note, a strong positive correlation was evident between the HMGB1 and that of TLR2 and TLR4 relative expression as well as IL-6 serum level. Conclusively, based on the observed changes in the analyzed markers in wound tissues and serum and R2 values obtained from mathematical models established to determine the wound age, the relative expression of HMGB1, TLR2, and TLR4 could be a reliable indicator for wound age determination in living subjects. Further investigation of these markers and mathematical models in human tissues is necessary.
Collapse
Affiliation(s)
- Yasmina M Abd-Elhakim
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt.
| | - Bothina H F Omran
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Human Medicine, Zagazig University, Zagazig, Egypt
| | - Shimaa A Ezzeldein
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Amany I Ahmed
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Nabela I El-Sharkawy
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Amany Abdel-Rahman Mohamed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt.
| |
Collapse
|
8
|
Kim J, Lee KM, Han SH, Ko EA, Yoon DS, Park IK, Shin HC, Park KH, Lee JW. Development of stabilized dual growth factor-loaded hyaluronate collagen dressing matrix. J Tissue Eng 2021; 12:2041731421999750. [PMID: 33796249 PMCID: PMC7970265 DOI: 10.1177/2041731421999750] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 02/13/2021] [Indexed: 12/26/2022] Open
Abstract
Patients with diabetes experience impaired growth factor production such as
epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF), and
they are reportedly involved in wound healing processes. Here, we report dual
growth factor-loaded hyaluronate collagen dressing (Dual-HCD) matrix, using
different ratios of the concentration of stabilized growth
factors—stabilized-EGF (S-EGF) and stabilized-bFGF (S-bFGF). At first, the
optimal concentration ratio of S-EGF to S-bFGF in the Dual-HCD matrix is
determined to be 1:2 in type I diabetic mice. This Dual-HCD matrix does not
cause cytotoxicity and can be used in vivo. The wound-healing effect of this
matrix is confirmed in type II diabetic mice. Dual HCD enhances angiogenesis
which promotes wound healing and thus, it shows a significantly greater
synergistic effect than the HCD matrix loaded with a single growth factor.
Overall, we conclude that the Dual-HCD matrix represents an effective
therapeutic agent for impaired diabetic wound healing.
Collapse
Affiliation(s)
- Jihyun Kim
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyoung-Mi Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung Hwan Han
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul, South Korea
| | - Eun Ae Ko
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Dong Suk Yoon
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Ik Kyu Park
- R&D Center, Genewel Co., Ltd., Sungnam, South Korea
| | - Hang-Cheol Shin
- School of Systems Biomedical Science, Soongsil University, Seoul, South Korea
| | - Kwang Hwan Park
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Jin Woo Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
9
|
Krzyszczyk P, Kang HJ, Kumar S, Meng Y, O’Reggio MD, Patel K, Pires IS, Yarmush ML, Schloss RS, Palmer AF, Berthiaume F. Anti-inflammatory effects of haptoglobin on LPS-stimulated macrophages: Role of HMGB1 signaling and implications in chronic wound healing. Wound Repair Regen 2020; 28:493-505. [PMID: 32428978 PMCID: PMC10927319 DOI: 10.1111/wrr.12814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 03/15/2020] [Accepted: 03/24/2020] [Indexed: 12/15/2022]
Abstract
Nonhealing wounds possess elevated numbers of pro-inflammatory M1 macrophages, which fail to transition to anti-inflammatory M2 phenotypes that promote healing. Hemoglobin (Hb) and haptoglobin (Hp) proteins, when complexed (Hb-Hp), can elicit M2-like macrophages through the heme oxygenase-1 (HO-1) pathway. Despite the fact that nonhealing wounds are chronically inflamed, previous studies have focused on non-inflammatory systems, and do not thoroughly compare the effects of complexed vs individual proteins. We aimed to investigate the effect of Hb/Hp treatments on macrophage phenotype in an inflammatory, lipopolysaccharide (LPS)-stimulated environment, similar to chronic wounds. Human M1 macrophages were cultured in vitro and stimulated with LPS. Concurrently, Hp, Hb, or Hb-Hp complexes were delivered. The next day, 27 proteins related to inflammation were measured in the supernatants. Hp treatment decreased a majority of inflammatory factors, Hb increased many, and Hb-Hp had intermediate trends, indicating that Hp attenuated overall inflammation to the greatest extent. From this data, Ingenuity Pathway Analysis software identified high motility group box 1 (HMGB1) as a key canonical pathway-strongly down-regulated from Hp, strongly up-regulated from Hb, and slightly activated from Hb-Hp. HMGB1 measurements in macrophage supernatants confirmed this trend. In vivo results in diabetic mice with biopsy punch wounds demonstrated accelerated wound closure with Hp treatment, and delayed wound closure with Hb treatment. This work specifically studied Hb/Hp effects on macrophages in a highly inflammatory environment relevant to chronic wound healing. Results show that Hp-and not Hb-Hp, which is known to be superior in noninflammatory conditions-reduces inflammation in LPS-stimulated macrophages, and HMGB1 signaling is also implicated. Overall, Hp treatment on M1 macrophages in vitro reduced the inflammatory secretion profile, and also exhibited benefits in in silico and in vivo wound-healing models.
Collapse
Affiliation(s)
- Paulina Krzyszczyk
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Hwan June Kang
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Yixin Meng
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Maurice D. O’Reggio
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Kishan Patel
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Ivan S. Pires
- William G. Lowrie Department of Chemical & Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Martin L. Yarmush
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Rene S. Schloss
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Andre F. Palmer
- William G. Lowrie Department of Chemical & Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - François Berthiaume
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
10
|
Alzokaky AAM, Abdelkader EM, El-Dessouki AM, Khaleel SA, Raslan NA. C-phycocyanin protects against ethanol-induced gastric ulcers in rats: Role of HMGB1/NLRP3/NF-κB pathway. Basic Clin Pharmacol Toxicol 2020; 127:265-277. [PMID: 32306544 DOI: 10.1111/bcpt.13415] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/27/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022]
Abstract
Gastric ulcer is a widespread inflammatory disease with high socio-economic burden. C-phycocyanin is one of the active constituents of Spirulina microalgae, and although it is well known for its antioxidant and anti-inflammatory properties, its protective effects against gastric ulcer have not yet been identified. High-mobility group box 1 (HMGB1) is a nuclear protein that, once secreted extracellularly, initiates several inflammatory reactions, and it is involved in the pathogenesis of gastric ulcer. The aim of the present study was to investigate the anti-inflammatory and anti-ulcerogenic effects of C-phycocyanin against ethanol-induced gastric ulcer targeting HMGB1/NLRP3/NF-κB pathway. Ulcer induction showed increase in HMGB1 expression through activation of nucleotide-binding domain and leucine-rich repeat-containing protein 3 (NLRP3) inflammasome and nuclear factor kappa p65 (NF-κB p65). Moreover, oxidative stress and inflammatory markers were elevated in the ulcer-treated group compared to the normal control group. However, pre-treatment with C-phycocyanin significantly reduced HMGB1 expression via suppression of NLRP3/NF-κB, oxidative markers, IL-1β, tumour necrosis factor-α (TNF-α) and ulcer index value. These results were consistent with histopathological and immunohistochemistry examination. Thus, C-phycocyanin is a potential therapeutic strategy with anti-inflammatory and anti-ulcerogenic effects against ethanol-induced gastric ulcer.
Collapse
Affiliation(s)
- Amany Abdel-Mageed Alzokaky
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University (Girls), Cairo, Egypt.,Pharmacology and biochemistry Department, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Eman M Abdelkader
- Department of Pharmacology & biochemistry, Faculty of Pharmacy, Delta University for Science &Technology, Mansoura, Egypt
| | - Ahmed M El-Dessouki
- Department of Pharmacology, Faculty of Pharmacy, Ahram Canadian University (ACU), Cairo, Egypt
| | - Sahar A Khaleel
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University (Girls), Cairo, Egypt.,Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Nahed A Raslan
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University (Girls), Cairo, Egypt.,Department of Emergency, AL-Ghad International College for applied medical Sciences, Jeddah, Saudi Arabia
| |
Collapse
|
11
|
Strohbuecker L, Koenen H, van Rijssen E, van Cranenbroek B, Fasse E, Joosten I, Körber A, Bergmann C. Increased dermal expression of chromatin-associated protein HMGB1 and concomitant T-cell expression of the DNA RAGE in patients with psoriasis vulgaris. PSORIASIS (AUCKLAND, N.Z.) 2019; 9:7-17. [PMID: 30859087 PMCID: PMC6385765 DOI: 10.2147/ptt.s190507] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Psoriasis vulgaris (PV) is an autoimmune-related chronic inflammatory disease of the skin, with both vascular and metabolic effects. Aggravating factors have been identified that initiate and maintain inflammation, including expression of Th1-, Th17-, and Th22-cell derived cytokines. Recently, we showed that the evolutionarily ancient and highly conserved damage-associated molecular pattern molecule "high mobility group box 1 (HMGB1)" is significantly increased in the serum of PV patients with disease progression and is decreased under standard therapies. MATERIALS AND METHODS To better understand the role of HMGB1 in the pathogenesis of PV, we recruited 22 untreated psoriatic patients with either mild or severe disease, defined by the Psoriasis Area Severity Index. We assessed HMGB1 and receptor for advanced glycation end products (RAGE) expression in the skin by immunohistochemistry and analyzed the immune-phenotype of Treg and Th17 cells by flow cytometry. RESULTS We found increased staining for HMGB1 in the dermis of psoriatic plaques in comparison to uninvolved skin of patients with PV. In addition, the major histocompatibility complex class III-encoded DNA and HMGB1 RAGE, induced by HMGB1, were highly expressed on psoriatic CD8+ T cells and CD4+ Treg. High expression of HMGB1 in the lesional skin was associated with even higher expression of its receptor, RAGE, on the cell surface of keratino-cytes in patients with severe PV. CONCLUSION The presence of HMGB1 and RAGE signaling may impact orchestration of chronic inflammation in PV which might have implications for Treg and Th17 cells.
Collapse
Affiliation(s)
- Lisa Strohbuecker
- Department of Dermatology, University Hospital Essen, 45147 Essen, Germany
| | - Hans Koenen
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Esther van Rijssen
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bram van Cranenbroek
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Esther Fasse
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Irma Joosten
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andreas Körber
- Department of Dermatology, University Hospital Essen, 45147 Essen, Germany
| | - Christoph Bergmann
- Department of Otorhinolaryngology, University Hospital Essen, 45147 Essen, Germany,
| |
Collapse
|
12
|
Zhang H, Lu X, Liu Z, Du K. Rosuvastatin reduces the pro-inflammatory effects of adriamycin on the expression of HMGB1 and RAGE in rats. Int J Mol Med 2018; 42:3415-3423. [PMID: 30320373 PMCID: PMC6202077 DOI: 10.3892/ijmm.2018.3928] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 10/03/2018] [Indexed: 12/22/2022] Open
Abstract
Rosuvastatin has cardiac protective effects through its anti‑inflammatory effects. The nuclear protein high‑mobility group box 1 (HMGB1) can activate inflammatory pathways when released from dying cells. The present study aimed to investigate the effects of rosuvastatin in adriamycin (ADR)‑treated rats. Adult male rats were randomized to three groups: i) Control group, ii) ADR group, and iii) ADR+rosuvastatin group. Serum biochemical indices were measured using an enzyme‑linked immunosorbent assay. Cardiac function was assessed by echocardiography. The expression of HMGB1 and receptors for advanced glycation end products (RAGE) were assessed by reverse transcription‑quantitative polymerase chain reaction analysis, western blot analysis, and immunohistochemistry. Cytokines were measured using flow cytometry. Rosuvastatin improved the biochemical indices and cardiac morphology and alleviated the pathological lesions. In the ADR+rosuvastatin group, the mRNA and protein levels of HMGB1 and RAGE in the myocardium were significantly lower compared with those in the ADR group (both P<0.05). The results showed that rosuvastatin significantly reduced the levels of HMGB1 and RAGE in the myocardium of the ADR‑treated rats. These results suggest that the protective effects of rosuvastatin may be associated with attenuation of the HMGB1/RAGE‑mediated inflammatory response in ADR‑treated rats. Despite this protective effect of rosuvastatin in the present study, it did not improve cardiac function in terms of the diastolic left ventricular internal dimension, systolic left ventricular internal dimension, left ventricular ejection fraction and left ventricular fractional shortening; this may be due the observation duration being insufficient.
Collapse
Affiliation(s)
- Haiyan Zhang
- Department of Cardiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xiang Lu
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zhengxia Liu
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Kang Du
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
13
|
Fournet M, Bonté F, Desmoulière A. Glycation Damage: A Possible Hub for Major Pathophysiological Disorders and Aging. Aging Dis 2018; 9:880-900. [PMID: 30271665 PMCID: PMC6147582 DOI: 10.14336/ad.2017.1121] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/21/2017] [Indexed: 12/25/2022] Open
Abstract
Glycation is both a physiological and pathological process which mainly affects proteins, nucleic acids and lipids. Exogenous and endogenous glycation produces deleterious reactions that take place principally in the extracellular matrix environment or within the cell cytosol and organelles. Advanced glycation end product (AGE) formation begins by the non-enzymatic glycation of free amino groups by sugars and aldehydes which leads to a succession of rearrangements of intermediate compounds and ultimately to irreversibly bound products known as AGEs. Epigenetic factors, oxidative stress, UV and nutrition are important causes of the accumulation of chemically and structurally different AGEs with various biological reactivities. Cross-linked proteins, deriving from the glycation process, present both an altered structure and function. Nucleotides and lipids are particularly vulnerable targets which can in turn favor DNA mutation or a decrease in cell membrane integrity and associated biological pathways respectively. In mitochondria, the consequences of glycation can alter bioenergy production. Under physiological conditions, anti-glycation defenses are sufficient, with proteasomes preventing accumulation of glycated proteins, while lipid turnover clears glycated products and nucleotide excision repair removes glycated nucleotides. If this does not occur, glycation damage accumulates, and pathologies may develop. Glycation-induced biological products are known to be mainly associated with aging, neurodegenerative disorders, diabetes and its complications, atherosclerosis, renal failure, immunological changes, retinopathy, skin photoaging, osteoporosis, and progression of some tumors.
Collapse
Affiliation(s)
- Maxime Fournet
- 1University of Limoges, Faculty of Pharmacy, Department of Physiology, EA 6309, F-87025 Limoges, France
| | | | - Alexis Desmoulière
- 3University of Limoges, Faculty of Pharmacy, Department of Physiology, EA 6309, F-87025 Limoges, France
| |
Collapse
|
14
|
Wilgus TA. Alerting the body to tissue injury: The role of alarmins and DAMPs in cutaneous wound healing. CURRENT PATHOBIOLOGY REPORTS 2018; 6:55-60. [PMID: 29862143 DOI: 10.1007/s40139-018-0162-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Purpose of review Tissue injury stimulates an inflammatory response that is mediated in part by alarmins. Alarmins are a group of endogenous molecules that trigger inflammation in response to damage. This class of molecules is becoming increasingly recognized for their ability to influence wound healing. This article will provide an overview of alarmins and outline the latest findings on these mediators in cutaneous wound healing. Recent findings In addition to stimulating inflammatory cells, recent evidence suggests that alarmins can act on other cells in the skin to affect wound closure and the extent of scar tissue production. This review will focus on HMGB-1 and IL-33, two alarmins that have received recent attention in the wound healing field. Summary Because a properly regulated inflammatory response is critical for optimal healing, further research must be done to fully understand the role of alarmins in the wound repair process.
Collapse
|
15
|
Nass N, Trau S, Paulsen F, Kaiser D, Kalinski T, Sel S. The receptor for advanced glycation end products RAGE is involved in corneal healing. Ann Anat 2017; 211:13-20. [DOI: 10.1016/j.aanat.2017.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 01/11/2023]
|
16
|
Khoo CP, Roubelakis MG, Schrader JB, Tsaknakis G, Konietzny R, Kessler B, Harris AL, Watt SM. miR-193a-3p interaction with HMGB1 downregulates human endothelial cell proliferation and migration. Sci Rep 2017; 7:44137. [PMID: 28276476 PMCID: PMC5343468 DOI: 10.1038/srep44137] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 02/02/2017] [Indexed: 12/12/2022] Open
Abstract
Circulating endothelial colony forming cells (ECFCs) contribute to vascular repair where they are a target for therapy. Since ECFC proliferative potential is increased in cord versus peripheral blood and to define regulatory factors controlling this proliferation, we compared the miRNA profiles of cord blood and peripheral blood ECFC-derived cells. Of the top 25 differentially regulated miRNAs selected, 22 were more highly expressed in peripheral blood ECFC-derived cells. After validating candidate miRNAs by q-RT-PCR, we selected miR-193a-3p for further investigation. The miR-193a-3p mimic reduced cord blood ECFC-derived cell proliferation, migration and vascular tubule formation, while the miR-193a-3p inhibitor significantly enhanced these parameters in peripheral blood ECFC-derived cells. Using in silico miRNA target database analyses combined with proteome arrays and luciferase reporter assays of miR-193a-3p mimic treated cord blood ECFC-derived cells, we identified 2 novel miR-193a-3p targets, the high mobility group box-1 (HMGB1) and the hypoxia upregulated-1 (HYOU1) gene products. HMGB1 silencing in cord blood ECFC-derived cells confirmed its role in regulating vascular function. Thus, we show, for the first time, that miR-193a-3p negatively regulates human ECFC vasculo/angiogenesis and propose that antagonising miR-193a-3p in less proliferative and less angiogenic ECFC-derived cells will enhance their vasculo/angiogenic function.
Collapse
Affiliation(s)
- Cheen P. Khoo
- Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9BQ, UK
- Stem Cell Research, NHS Blood and Transplant, Oxford, OX3 9BQ, UK
| | - Maria G. Roubelakis
- Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9BQ, UK
- Stem Cell Research, NHS Blood and Transplant, Oxford, OX3 9BQ, UK
- Laboratory of Biology, National and Kapodistrian University of Athens Medical School, Athens 115 27, Greece
- Cell and Gene Therapy Laboratory, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, 11527, Greece
| | - Jack B. Schrader
- Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9BQ, UK
- Stem Cell Research, NHS Blood and Transplant, Oxford, OX3 9BQ, UK
- Department of Biology, University of York, York, YO10 5DD, UK
| | - Grigorios Tsaknakis
- Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9BQ, UK
- Stem Cell Research, NHS Blood and Transplant, Oxford, OX3 9BQ, UK
- Institute of Molecular Biology and Biotechnology, Foundation of Research & Technology, GR-70013 Heraklion, Crete
| | - Rebecca Konietzny
- Target Discovery Institute, NDM Research Building, Nuffield Department of Medicine, University of Oxford, OX3 7FZ, UK
| | - Benedikt Kessler
- Target Discovery Institute, NDM Research Building, Nuffield Department of Medicine, University of Oxford, OX3 7FZ, UK
| | - Adrian L. Harris
- The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Suzanne M. Watt
- Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9BQ, UK
- Stem Cell Research, NHS Blood and Transplant, Oxford, OX3 9BQ, UK
| |
Collapse
|
17
|
Van Putte L, De Schrijver S, Moortgat P. The effects of advanced glycation end products (AGEs) on dermal wound healing and scar formation: a systematic review. Scars Burn Heal 2016; 2:2059513116676828. [PMID: 29799552 PMCID: PMC5965313 DOI: 10.1177/2059513116676828] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Introduction: With ageing, the skin gradually loses its youthful appearance and functions
like wound healing and scar formation. The pathophysiological theory of
Advanced Glycation End products (AGEs) has gained traction during the last
decade. This review aims to document the influence of AGEs on the mechanical
and physiologic properties of the skin, how they affect dermal wound healing
and scar formation in high-AGE populations like elderly patients and
diabetics, and potential therapeutic strategies. Methods: This systematic literature study involved a structured search in Pubmed and
Web of Science with qualitative analysis of 14 articles after a three-staged
selection process with the use of in- and exclusion criteria. Results: Overall, AGEs cause shortened, thinned, and disorganized collagen fibrils,
consequently reducing elasticity and skin/scar thickness with increased
contraction and delayed wound closure. Documented therapeutic strategies
include dietary AGE restriction, sRAGE decoy receptors, aminoguanidine,
RAGE-blocking antibodies, targeted therapy, thymosin β4, anti-oxidant agents
and gold nanoparticles, ethyl pyruvate, Gal-3 manipulation and
metformin. Discussion: With lack of evidence concerning scars, no definitive conclusions can yet be
made about the role of AGEs on possible appearance or function of scar
tissue. However, all results suggest that scars tend to be more rigid and
contractile with persistent redness and reduced tendency towards hypertrophy
as AGEs accumulate. Conclusion: Abundant evidence supports the pathologic role of AGEs in ageing and dermal
wound healing and the effectiveness of possible therapeutic agents. More
research is required to conclude its role in scar formation and scar
therapy. Our skin is the body’s first line of defense. It is the barrier that protects us
from chemical and biological threats such as viruses, bacteria or corrosive
liquids. It is the sensor that allows us to detect physical threats like extreme
temperatures, pressure and pain. And when these preventative measures fail, the
skin has yet another property: the ability to heal. Skin changes visibly with age, most notably with the appearance of wrinkles.
However, there is more to ageing than meets the eye; invisible alterations cause
the decline of various functions of the skin, such as wound healing and scar
formation. An array of non-conclusive research has been done in this field. One
theory that has gained traction during the last decade is the Advanced Glycation
End products (AGEs) theory. The theory states that AGEs play an important role
in skin aging, wound healing and the effectiveness of different therapeutic
options. Their presence supposedly indicates a diminished ability for wound
healing and scar formation. AGEs are proteins to which sugar molecule is bound. The sugar molecule inhibits
the original protein from functioning properly. As skin contains many proteins
like collagen, the formation of these AGEs could be a viable explanation for the
diminished functioning with ageing. In this review, we investigated whether the
accumulation of AGEs affects wound healing and scar formation. Normal scar formation results in a thin scar. However, it may happen that
scarring results in thick, large, painful and itchy scars. We investigated
whether people with a high AGE content in their skin, like diabetics and
elderly, have difficulties forming aesthetically pleasing scars. Secondly, we
investigated which therapies reduce the AGE content and, if so, whether these
therapies can improve wound healing and scarring. This literature study involved
research in scientific databases with qualitative analysis of 14 articles after
a three-staged selection process with the use of set criteria. We found the different ways in which AGEs affect skin properties and wound
healing. Collagen, one of the most important proteins in the skin, is affected
by these AGEs. Once a sugar binds to it, the collagen strings becomes thinner
and shorter, and the different collagen proteins cross-link with each other in
an unstructured way. The result of these alterations is a reduced elasticity,
i.e. the skin becomes stiffer. The scar will be thinner and the time for wounds
to close is longer. We also found strategies to diminish the AGE content,
including dietary AGE restriction and Metformin, a drug used in diabetes. We can conclude that there is proof of AGEs playing an important role in skin
ageing, wound healing and the effectiveness of different therapeutic options.
However, more research is required to conclude the exact role of AGEs in scar
formation and scar therapy.
Collapse
Affiliation(s)
- Lennert Van Putte
- Faculty of Medicine and Health Science, University of Antwerp, Antwerp, Belgium
| | - Sofie De Schrijver
- Faculty of Medicine and Health Science, University of Antwerp, Antwerp, Belgium
| | - Peter Moortgat
- Oscare, Organisation for Burns, Scar After-care and Research, Antwerp, Belgium
| |
Collapse
|
18
|
Tancharoen S, Gando S, Binita S, Nagasato T, Kikuchi K, Nawa Y, Dararat P, Yamamoto M, Narkpinit S, Maruyama I. HMGB1 Promotes Intraoral Palatal Wound Healing through RAGE-Dependent Mechanisms. Int J Mol Sci 2016; 17:ijms17111961. [PMID: 27886093 PMCID: PMC5133955 DOI: 10.3390/ijms17111961] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/05/2016] [Accepted: 11/15/2016] [Indexed: 12/24/2022] Open
Abstract
High mobility group box 1 (HMGB1) is tightly connected to the process of tissue organization upon tissue injury. Here we show that HMGB1 controls epithelium and connective tissue regeneration both in vivo and in vitro during palatal wound healing. Heterozygous HMGB1 (Hmgb1+/-) mice and Wild-type (WT) mice were subjected to palatal injury. Maxillary tissues were stained with Mallory Azan or immunostained with anti-HMGB1, anti-proliferating cell nuclear antigen (PCNA), anti-nuclear factor-κB (NF-κB) p50 and anti-vascular endothelial growth factor (VEGF) antibodies. Palatal gingival explants were cultured with recombinant HMGB1 (rHMGB1) co-treated with siRNA targeting receptor for advanced glycation end products (RAGEs) for cell migration and PCNA expression analysis. Measurement of the wound area showed differences between Hmgb1+/- and WT mice on Day 3 after wounding. Mallory Azan staining showed densely packed of collagen fibers in WT mice, whereas in Hmgb1+/- mice weave-like pattern of low density collagen bundles were present. At three and seven days post-surgery, PCNA, NF-κB p50 and VEGF positive keratinocytes of WT mice were greater than that of Hmgb1+/- mice. Knockdown of RAGE prevents the effect of rHMGB1-induced cell migration and PCNA expression in gingival cell cultures. The data suggest that HMGB1/RAGE axis has crucial roles in palatal wound healing.
Collapse
Affiliation(s)
- Salunya Tancharoen
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand.
| | - Satoshi Gando
- Department of Emergency and Critical Care, Hokkaido University, Kita-ku, Sapporo 0608648, Japan.
| | - Shrestha Binita
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 8908544, Japan.
| | - Tomoka Nagasato
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 8908544, Japan.
| | - Kiyoshi Kikuchi
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand.
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, Asahi-machi, Kurume 8300011, Japan.
| | - Yuko Nawa
- Department of Anesthesiology and Intensive Care, Hokkaido Medical for Child Health and Rehabilitation, Sapporo 0060041, Japan.
| | - Pornpen Dararat
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand.
| | - Mika Yamamoto
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 8908544, Japan.
| | - Somphong Narkpinit
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| | - Ikuro Maruyama
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 8908544, Japan.
| |
Collapse
|
19
|
Pandolfi F, Altamura S, Frosali S, Conti P. Key Role of DAMP in Inflammation, Cancer, and Tissue Repair. Clin Ther 2016; 38:1017-28. [PMID: 27021609 DOI: 10.1016/j.clinthera.2016.02.028] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 02/29/2016] [Accepted: 02/29/2016] [Indexed: 12/30/2022]
Abstract
PURPOSE This review aimed to take stock of the current status of research on damage-associated molecular pattern (DAMP) protein. We discuss the Janus-faced role of DAMP molecules in inflammation, cancer, and tissue repair. The high-mobility group box (HMGB)-1 and adenosine triphosphate proteins are well-known DAMP molecules and have been primarily associated with inflammation. However, as we shall see, recent data have linked these molecules to tissue repair. HMGB1 is associated with cancer-related inflammation. It activates nuclear factor kB, which is involved in cancer regulation via its receptor for advanced glycation end-products (RAGE), Toll-like receptors 2 and 4. Proinflammatory activity and tissue repair may lead to pharmacologic intervention, by blocking DAMP RAGE and Toll like receptor 2 and 4 role in inflammation and by increasing their concentration in tissue repair, respectively. METHODS We conducted a MEDLINE search for articles pertaining to the various issues related to DAMP, and we discuss the most relevant articles especially (ie, not only those published in journals with a higher impact factor). FINDINGS A cluster of remarkable articles on DAMP have appeared in the literature in recent years. Regarding inflammation, several strategies have been proposed to target HMGB1, from antibodies to recombinant box A, which interacts with RAGE, competing with the full molecule. In tissue repair, it was reported that the overexpression of HMGB1 or the administration of exogenous HMGB1 significantly increased the number of vessels and promoted recovery in skin-wound, ischemic injury. IMPLICATIONS Due to the bivalent nature of DAMP, it is often difficult to explain the relative role of DAMP in inflammation versus its role in tissue repair. However, this point is crucial as DAMP-related treatments move into clinical practice.
Collapse
Affiliation(s)
- Franco Pandolfi
- Department of Internal Medicine, School of Medicine, Catholic University, Rome, Italy.
| | - Simona Altamura
- Department of Internal Medicine, School of Medicine, Catholic University, Rome, Italy
| | - Simona Frosali
- Department of Internal Medicine, School of Medicine, Catholic University, Rome, Italy
| | - Pio Conti
- Postgraduate Medical School, Chieti University, Chieti, Italy
| |
Collapse
|
20
|
Starzl R, Wolfram D, Zamora R, Jefferson B, Barclay D, Ho C, Gorantla V, Brandacher G, Schneeberger S, Andrew Lee WP, Carbonell J, Vodovotz Y. Cardiac Arrest Disrupts Caspase-1 and Patterns of Inflammatory Mediators Differently in Skin and Muscle Following Localized Tissue Injury in Rats: Insights from Data-Driven Modeling. Front Immunol 2015; 6:587. [PMID: 26635801 PMCID: PMC4653302 DOI: 10.3389/fimmu.2015.00587] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 11/02/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Trauma often cooccurs with cardiac arrest and hemorrhagic shock. Skin and muscle injuries often lead to significant inflammation in the affected tissue. The primary mechanism by which inflammation is initiated, sustained, and terminated is cytokine-mediated immune signaling, but this signaling can be altered by cardiac arrest. The complexity and context sensitivity of immune signaling in general has stymied a clear understanding of these signaling dynamics. METHODOLOGY/PRINCIPAL FINDINGS We hypothesized that advanced numerical and biological function analysis methods would help elucidate the inflammatory response to skin and muscle wounds in rats, both with and without concomitant shock. Based on the multiplexed analysis of inflammatory mediators, we discerned a differential interleukin (IL)-1α and IL-18 signature in skin vs. muscle, which was suggestive of inflammasome activation in the skin. Immunoblotting revealed caspase-1 activation in skin but not muscle. Notably, IL-1α and IL-18, along with caspase-1, were greatly elevated in the skin following cardiac arrest, consistent with differential inflammasome activation. CONCLUSION/SIGNIFICANCE Tissue-specific activation of caspase-1 and the NLRP3 inflammasome appear to be key factors in determining the type and severity of the inflammatory response to tissue injury, especially in the presence of severe shock, as suggested via data-driven modeling.
Collapse
Affiliation(s)
- Ravi Starzl
- Language Technologies Institute, Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Plastic and Reconstructive Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Plastic and Reconstructive Surgery, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Dolores Wolfram
- Department of Plastic and Reconstructive Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - Ruben Zamora
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Derek Barclay
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chien Ho
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Vijay Gorantla
- Department of Plastic and Reconstructive Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gerald Brandacher
- Department of Plastic and Reconstructive Surgery, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Stefan Schneeberger
- Department of Plastic and Reconstructive Surgery, Johns Hopkins Medicine, Baltimore, MD, USA
| | - W. P. Andrew Lee
- Department of Plastic and Reconstructive Surgery, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Jaime Carbonell
- Language Technologies Institute, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
21
|
Tsai SC, Jheng YH, Wang CY, Chen YW, Lin YF, Chen CC, Chang PC. Osseous wound repair under inhibition of the axis of advanced glycation end-products and the advanced glycation end-products receptor. J Formos Med Assoc 2015; 114:973-80. [DOI: 10.1016/j.jfma.2013.11.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/06/2013] [Accepted: 11/27/2013] [Indexed: 10/25/2022] Open
|
22
|
Karuppagounder V, Arumugam S, Thandavarayan RA, Pitchaimani V, Sreedhar R, Afrin R, Harima M, Suzuki H, Nomoto M, Miyashita S, Suzuki K, Watanabe K. Resveratrol attenuates HMGB1 signaling and inflammation in house dust mite-induced atopic dermatitis in mice. Int Immunopharmacol 2015; 23:617-23. [PMID: 25466270 DOI: 10.1016/j.intimp.2014.10.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 10/16/2014] [Accepted: 10/16/2014] [Indexed: 01/22/2023]
Abstract
Resveratrol is a polyphenol abundantly found in red grape skin and is effective against antiaging and anti-inflammation associated with immune responses. In this study, we have investigated the effect of resveratrol on skin lesion, high mobility group box (HMGB)1 and inflammation pathway in an atopic dermatitis (AD) mouse model. AD-like lesion was induced by the application of house dust mite extract to the dorsal skin of NC/Nga mouse. After AD induction, resveratrol (20 mg/kg, p.o.) was administered daily for 2 weeks. We evaluated dermatitis severity, histopathological changes, serum levels of T helper (Th) cytokines (interferon (IFN)γ, interleukin (IL)-4) and changes in protein expression by Western blotting for HMGB1, receptor for advanced glycation end products (RAGE), toll like receptor (TLR)4, nuclear factor (NF)κB, phosphatidylinositide 3-kinase (PI3K), extracellular signal-regulated kinase (ERK)1/2, cyclooxygenase (COX)2, tumor necrosis factor (TNF)α, IL-1β, IL-2Rα and other inflammatory markers in the skin of AD mice. Treatment of resveratrol inhibited the development of the AD-like skin lesions. Histological analysis showed that resveratrol inhibited hypertrophy, intracellular edema, mast cells and infiltration of inflammatory cells. Furthermore, resveratrol treatment down-regulated HMGB1, RAGE, p-NFκB, p-PI3K, p-ERK1/2, COX2, TNFα, IL-1β, IL-2Rα, IFNγ and IL-4. Considering all these findings together, the HMGB1 pathway might be a potential therapeutic target in skin inflammation, and resveratrol treatment could have beneficial effects on AD by modulating the HMGB1 protein expression.
Collapse
|
23
|
Innate Immunity and Biomaterials at the Nexus: Friends or Foes. BIOMED RESEARCH INTERNATIONAL 2015; 2015:342304. [PMID: 26247017 PMCID: PMC4515263 DOI: 10.1155/2015/342304] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 06/15/2015] [Accepted: 06/22/2015] [Indexed: 01/04/2023]
Abstract
Biomaterial implants are an established part of medical practice, encompassing a broad range of devices that widely differ in function and structural composition. However, one common property amongst biomaterials is the induction of the foreign body response: an acute sterile inflammatory reaction which overlaps with tissue vascularisation and remodelling and ultimately fibrotic encapsulation of the biomaterial to prevent further interaction with host tissue. Severity and clinical manifestation of the biomaterial-induced foreign body response are different for each biomaterial, with cases of incompatibility often associated with loss of function. However, unravelling the mechanisms that progress to the formation of the fibrotic capsule highlights the tightly intertwined nature of immunological responses to a seemingly noncanonical “antigen.” In this review, we detail the pathways associated with the foreign body response and describe possible mechanisms of immune involvement that can be targeted. We also discuss methods of modulating the immune response by altering the physiochemical surface properties of the biomaterial prior to implantation. Developments in these areas are reliant on reproducible and effective animal models and may allow a “combined” immunomodulatory approach of adapting surface properties of biomaterials, as well as treating key immune pathways to ultimately reduce the negative consequences of biomaterial implantation.
Collapse
|
24
|
Lee DE, Trowbridge RM, Ayoub NT, Agrawal DK. High-mobility Group Box Protein-1, Matrix Metalloproteinases, and Vitamin D in Keloids and Hypertrophic Scars. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2015; 3:e425. [PMID: 26180726 PMCID: PMC4494495 DOI: 10.1097/gox.0000000000000391] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/23/2015] [Indexed: 01/16/2023]
Abstract
Keloids and hypertrophic scars represent excessive wound healing involving high production of collagen by skin fibroblasts. This review focuses on the role of high-mobility group box protein-1 (HMGB-1), matrix metalloproteinases (MMPs), and vitamin D in these conditions. Although the role of HMGB-1 in keloids and hypertrophic scars is unclear, the effect of HMGB-1 on fibroblasts suggests a profibrotic role and a potential contribution to excessive scarring. MMPs contribute extensively to wound healing and characteristically degrade the extracellular matrix. MMP-1 is decreased in keloids and hypertrophic scars. However, other MMPs, including MMP-2, have been found to be increased and are thought to possibly contribute to keloid expansion through peripheral extracellular matrix catabolism. Many novel therapeutic approaches to keloids and hypertrophic scars target MMPs and aim to increase their levels and catabolic activity. The higher prevalence of keloids in darker skin types may partially be due to a tendency for lower vitamin D levels. The physiologically active form of vitamin D, 1,25(OH)2D3, inhibits the proliferation of keloid fibroblasts, and correlations between vitamin D receptor polymorphisms, such as the TaqI CC genotype, and keloid formation have been reported. Additionally, vitamin D may exert an antifibrotic effect partially mediated by MMPs. Here, we critically discuss whether keloid and hypertrophic scar formation could be predicted based on vitamin D status and vitamin D receptor polymorphisms. Specifically, the findings identified HMGB-1, MMPs, and vitamin D as potential avenues for further clinical investigation and potentially novel therapeutic approaches to prevent the development of keloids and hypertrophic scars.
Collapse
Affiliation(s)
- Dylan E. Lee
- From the Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Neb.; Brigham and Women’s Hospital, Boston, Mass.; and Westfield Plastic Surgery Center, Omaha, Neb
| | - Ryan M. Trowbridge
- From the Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Neb.; Brigham and Women’s Hospital, Boston, Mass.; and Westfield Plastic Surgery Center, Omaha, Neb
| | - Nagi T. Ayoub
- From the Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Neb.; Brigham and Women’s Hospital, Boston, Mass.; and Westfield Plastic Surgery Center, Omaha, Neb
| | - Devendra K. Agrawal
- From the Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Neb.; Brigham and Women’s Hospital, Boston, Mass.; and Westfield Plastic Surgery Center, Omaha, Neb
| |
Collapse
|
25
|
Godefroy E, Gallois A, Idoyaga J, Merad M, Tung N, Monu N, Saenger Y, Fu Y, Ravindran R, Pulendran B, Jotereau F, Trombetta S, Bhardwaj N. Activation of toll-like receptor-2 by endogenous matrix metalloproteinase-2 modulates dendritic-cell-mediated inflammatory responses. Cell Rep 2014; 9:1856-1870. [PMID: 25466255 PMCID: PMC4336179 DOI: 10.1016/j.celrep.2014.10.067] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 09/22/2014] [Accepted: 10/29/2014] [Indexed: 10/24/2022] Open
Abstract
Matrix metalloproteinase-2 (MMP-2) is involved in several physiological mechanisms, including wound healing and tumor progression. We show that MMP-2 directly stimulates dendritic cells (DCs) to both upregulate OX40L on the cell surface and secrete inflammatory cytokines. The mechanism underlying DC activation includes physical association with Toll-like receptor-2 (TLR2), leading to NF-κB activation, OX40L upregulation on DCs, and ensuing TH2 differentiation. Significantly, MMP-2 polarizes T cells toward type 2 responses in vivo, in a TLR2-dependent manner. MMP-2-dependent type 2 polarization may represent a key immune regulatory mechanism for protection against a broad array of disorders, such as inflammatory, infectious, and autoimmune diseases, which can be hijacked by tumors to evade immunity.
Collapse
Affiliation(s)
- Emmanuelle Godefroy
- Mount Sinai School of Medicine, Tisch Cancer Institute, New York, NY 10029, USA
| | - Anne Gallois
- Mount Sinai School of Medicine, Tisch Cancer Institute, New York, NY 10029, USA
| | - Juliana Idoyaga
- Mount Sinai School of Medicine, Tisch Cancer Institute, New York, NY 10029, USA; Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, New York, NY 10065, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Miriam Merad
- Mount Sinai School of Medicine, Tisch Cancer Institute, New York, NY 10029, USA
| | - Navpreet Tung
- Mount Sinai School of Medicine, Tisch Cancer Institute, New York, NY 10029, USA
| | - Ngozi Monu
- New York University Langone Medical Center, Cancer Institute, New York, NY 10016, USA
| | - Yvonne Saenger
- Mount Sinai School of Medicine, Tisch Cancer Institute, New York, NY 10029, USA
| | - Yichun Fu
- Mount Sinai School of Medicine, Tisch Cancer Institute, New York, NY 10029, USA
| | - Rajesh Ravindran
- Emory Vaccine Center, Department of Pathology, Emory University, Atlanta, GA 30322, USA
| | - Bali Pulendran
- Emory Vaccine Center, Department of Pathology, Emory University, Atlanta, GA 30322, USA
| | | | - Sergio Trombetta
- New York University Langone Medical Center, Cancer Institute, New York, NY 10016, USA
| | - Nina Bhardwaj
- Mount Sinai School of Medicine, Tisch Cancer Institute, New York, NY 10029, USA.
| |
Collapse
|
26
|
Anabolic properties of high mobility group box protein-1 in human periodontal ligament cells in vitro. Mediators Inflamm 2014; 2014:347585. [PMID: 25525297 PMCID: PMC4265691 DOI: 10.1155/2014/347585] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 08/18/2014] [Indexed: 01/10/2023] Open
Abstract
High mobility group box protein-1 (HMGB1) is mainly recognized as a chemoattractant for macrophages in the initial phase of host response to pathogenic stimuli. However, recent findings provide evidence for anabolic properties in terms of enhanced proliferation, migration, and support of wound healing capacity of mesenchymal cells suggesting a dual role of the cytokine in the regulation of immune response and subsequent regenerative processes. Here, we examined potential anabolic effects of HMGB1 on human periodontal ligament (PDL) cells in the regulation of periodontal remodelling, for example, during orthodontic tooth movement. Preconfluent human PDL cells (hPDL) were exposed to HMGB1 protein and the influence on proliferation, migration, osteogenic differentiation, and biomineralization was determined by MTS assay, real time PCR, immunofluorescence cytochemistry, ELISA, and von Kossa staining. HMGB1 protein increased hPDL cell proliferation, migration, osteoblastic marker gene expression, and protein production as well as mineralized nodule formation significantly. The present findings support the dual character of HMGB1 with anabolic therapeutic potential that might support the reestablishment of the structural and functional integrity of the periodontium following periodontal trauma such as orthodontic tooth movement.
Collapse
|
27
|
Potter KA, Jorfi M, Householder KT, Foster EJ, Weder C, Capadona JR. Curcumin-releasing mechanically adaptive intracortical implants improve the proximal neuronal density and blood-brain barrier stability. Acta Biomater 2014; 10:2209-22. [PMID: 24468582 DOI: 10.1016/j.actbio.2014.01.018] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 12/09/2013] [Accepted: 01/15/2014] [Indexed: 10/25/2022]
Abstract
The cellular and molecular mechanisms by which neuroinflammatory pathways respond to and propagate the reactive tissue response to intracortical microelectrodes remain active areas of research. We previously demonstrated that both the mechanical mismatch between rigid implants and the much softer brain tissue, as well as oxidative stress, contribute to the neurodegenerative reactive tissue response to intracortical implants. In this study, we utilize physiologically responsive, mechanically adaptive polymer implants based on poly(vinyl alcohol) (PVA), with the capability to also locally administer the antioxidant curcumin. The goal of this study is to investigate if the combination of two independently effective mechanisms - softening of the implant and antioxidant release - leads to synergistic effects in vivo. Over the first 4weeks of the implantation, curcumin-releasing, mechanically adaptive implants were associated with higher neuron survival and a more stable blood-brain barrier at the implant-tissue interface than the neat PVA controls. 12weeks post-implantation, the benefits of the curcumin release were lost, and both sets of compliant materials (with and without curcumin) had no statistically significant differences in neuronal density distribution profiles. Overall, however, the curcumin-releasing softening polymer implants cause minimal implant-mediated neuroinflammation, and embody the new concept of localized drug delivery from mechanically adaptive intracortical implants.
Collapse
|
28
|
Chang PC, Tsai SC, Jheng YH, Lin YF, Chen CC. Soft-tissue wound healing by anti-advanced glycation end-products agents. J Dent Res 2014; 93:388-93. [PMID: 24554538 DOI: 10.1177/0022034514523785] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The blocking of advanced glycation end-products (AGE) has been shown to reduce diabetic complications and control periodontitis. This study investigated the pattern of palatal wound-healing after graft harvesting under the administration of aminoguanidine (AG), an AGE inhibitor, or N-phenacylthiazolium bromide (PTB), a glycated cross-link breaker. Full-thickness palatal excisional wounds (5.0 x 1.5 mm(2)) were created in 72 Sprague-Dawley rats. The rats received daily intraperitoneal injections of normal saline (control), AG, or PTB and were euthanized after 4 to 28 days. The wound-healing pattern was assessed by histology, histochemistry for collagen matrix deposition, immunohistochemistry for AGE and the AGE receptor (RAGE), and the expression of RAGE, as well as inflammation- and recovery-associated genes. In the first 14 days following AG or PTB treatments, wound closure, re-epithelialization, and collagen matrix deposition were accelerated, whereas AGE deposition, RAGE-positive cells, and inflammation were reduced. RAGE and tumor necrosis factor-alpha were significantly down-regulated at day 7, and heme oxygenase-1 was persistently down-regulated until day 14. The levels of vascular endothelial growth factor, periostin, type I collagen, and fibronectin were all increased at day 14. In conclusion, anti-AGE agents appeared to facilitate palatal wound-healing by reducing AGE-associated inflammation and promoting the recovery process.
Collapse
Affiliation(s)
- P-C Chang
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
29
|
High-mobility group box 1 inhibits gastric ulcer healing through Toll-like receptor 4 and receptor for advanced glycation end products. PLoS One 2013; 8:e80130. [PMID: 24244627 PMCID: PMC3823709 DOI: 10.1371/journal.pone.0080130] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 09/30/2013] [Indexed: 01/17/2023] Open
Abstract
High-mobility group box 1 (HMGB1) was initially discovered as a nuclear protein that interacts with DNA as a chromatin-associated non-histone protein to stabilize nucleosomes and to regulate the transcription of many genes in the nucleus. Once leaked or actively secreted into the extracellular environment, HMGB1 activates inflammatory pathways by stimulating multiple receptors, including Toll-like receptor (TLR) 2, TLR4, and receptor for advanced glycation end products (RAGE), leading to tissue injury. Although HMGB1’s ability to induce inflammation has been well documented, no studies have examined the role of HMGB1 in wound healing in the gastrointestinal field. The aim of this study was to evaluate the role of HMGB1 and its receptors in the healing of gastric ulcers. We also investigated which receptor among TLR2, TLR4, or RAGE mediates HMGB1’s effects on ulcer healing. Gastric ulcers were induced by serosal application of acetic acid in mice, and gastric tissues were processed for further evaluation. The induction of ulcer increased the immunohistochemical staining of cytoplasmic HMGB1 and elevated serum HMGB1 levels. Ulcer size, myeloperoxidase (MPO) activity, and the expression of tumor necrosis factor α (TNFα) mRNA peaked on day 4. Intraperitoneal administration of HMGB1 delayed ulcer healing and elevated MPO activity and TNFα expression. In contrast, administration of anti-HMGB1 antibody promoted ulcer healing and reduced MPO activity and TNFα expression. TLR4 and RAGE deficiency enhanced ulcer healing and reduced the level of TNFα, whereas ulcer healing in TLR2 knockout (KO) mice was similar to that in wild-type mice. In TLR4 KO and RAGE KO mice, exogenous HMGB1 did not affect ulcer healing and TNFα expression. Thus, we showed that HMGB1 is a complicating factor in the gastric ulcer healing process, which acts through TLR4 and RAGE to induce excessive inflammatory responses.
Collapse
|
30
|
Aldini G, Vistoli G, Stefek M, Chondrogianni N, Grune T, Sereikaite J, Sadowska-Bartosz I, Bartosz G. Molecular strategies to prevent, inhibit, and degrade advanced glycoxidation and advanced lipoxidation end products. Free Radic Res 2013; 47 Suppl 1:93-137. [PMID: 23560617 DOI: 10.3109/10715762.2013.792926] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advanced glycoxidation end products (AGEs) and lipoxidation end products (ALEs) contribute to the development of diabetic complications and of other pathologies. The review discusses the possibilities of counteracting the formation and stimulating the degradation of these species by pharmaceuticals and natural compounds. The review discusses inhibitors of ALE and AGE formation, cross-link breakers, ALE/AGE elimination by enzymes and proteolytic systems, receptors for advanced glycation end products (RAGEs) and blockade of the ligand-RAGE axis.
Collapse
Affiliation(s)
- Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Dardenne AD, Wulff BC, Wilgus TA. The alarmin HMGB-1 influences healing outcomes in fetal skin wounds. Wound Repair Regen 2013; 21:282-91. [PMID: 23438257 DOI: 10.1111/wrr.12028] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 12/25/2012] [Indexed: 01/05/2023]
Abstract
In mice, cutaneous wounds generated early in development (embryonic day 15, E15) heal scarlessly, while wounds generated late in gestation (embryonic day 18, E18) heal with scar formation. Even though both types of wounds are generated in the same sterile uterine environment, scarless fetal wounds heal without inflammation, but a strong inflammatory response is observed in scar-forming fetal wounds. We hypothesized that altered release of alarmins, endogenous molecules that trigger inflammation in response to damage, may be responsible for the age-related changes in inflammation and healing outcomes in fetal skin. The purpose of this study was to determine whether the alarmin high-mobility group box-1 (HMGB-1) is involved in fetal wound repair. Immunohistochemical analysis showed that in unwounded skin, E18 keratinocytes expressed higher levels of HMGB-1 compared with E15 keratinocytes. After injury, HMGB-1 was released to a greater extent from keratinocytes at the margin of scar-forming E18 wounds, compared with scarless E15 wounds. Furthermore, instead of healing scarlessly, E15 wounds healed with scars when treated with HMGB-1. HMGB-1-injected wounds also had more fibroblasts, blood vessels, and macrophages compared with control wounds. Together, these data suggest that extracellular HMGB-1 levels influence the quality of healing in cutaneous wounds.
Collapse
Affiliation(s)
- Adrienne D Dardenne
- Department of Veterinary Preventive Medicine, University Laboratory Animal Resources, The Ohio State University, Columbus, OH, USA
| | | | | |
Collapse
|
32
|
Seol D, McCabe DJ, Choe H, Zheng H, Yu Y, Jang K, Walter MW, Lehman AD, Ding L, Buckwalter JA, Martin JA. Chondrogenic progenitor cells respond to cartilage injury. ACTA ACUST UNITED AC 2013; 64:3626-3637. [PMID: 22777600 DOI: 10.1002/art.34613] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Hypocellularity resulting from chondrocyte death in the aftermath of mechanical injury is thought to contribute to posttraumatic osteoarthritis. However, we observed that nonviable areas in cartilage injured by blunt impact were repopulated within 7-14 days by cells that appeared to migrate from the surrounding matrix. The aim of this study was to assess our hypothesis that the migrating cell population included chondrogenic progenitor cells that were drawn to injured cartilage by alarmins. METHODS Osteochondral explants obtained from mature cattle were injured by blunt impact or scratching, resulting in localized chondrocyte death. Injured sites were serially imaged by confocal microscopy, and migrating cells were evaluated for chondrogenic progenitor characteristics. Chemotaxis assays were used to measure the responses to chemokines, injury-conditioned medium, dead cell debris, and high mobility group box chromosomal protein 1 (HMGB-1). RESULTS Migrating cells were highly clonogenic and multipotent and expressed markers associated with chondrogenic progenitor cells. Compared with chondrocytes, these cells overexpressed genes involved in proliferation and migration and underexpressed cartilage matrix genes. They were more active than chondrocytes in chemotaxis assays and responded to cell lysates, conditioned medium, and HMGB-1. Glycyrrhizin, a chelator of HMGB-1 and a blocking antibody to receptor for advanced glycation end products (RAGE), inhibited responses to cell debris and conditioned medium and reduced the numbers of migrating cells on injured explants. CONCLUSION Injuries that caused chondrocyte death stimulated the emergence and homing of chondrogenic progenitor cells, in part via HMGB-1 release and RAGE-mediated chemotaxis. Their repopulation of the matrix could promote the repair of chondral damage that might otherwise contribute to progressive cartilage loss.
Collapse
Affiliation(s)
- Dongrim Seol
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Daniel J McCabe
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Hyeonghun Choe
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Hongjun Zheng
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Yin Yu
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Keewoong Jang
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Morgan W Walter
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Abigail D Lehman
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Lei Ding
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Joseph A Buckwalter
- Joseph A. Buckwalter, MD: University of Iowa and VA Medical Center, Iowa City, Iowa
| | - James A Martin
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| |
Collapse
|
33
|
Ranzato E, Martinotti S, Pedrazzi M, Patrone M. High mobility group box protein-1 in wound repair. Cells 2012; 1:699-710. [PMID: 24710526 PMCID: PMC3901153 DOI: 10.3390/cells1040699] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 09/10/2012] [Accepted: 09/19/2012] [Indexed: 12/21/2022] Open
Abstract
High-mobility group box 1 protein (HMGB1), a member of highly conserved non-histone DNA binding protein family, has been studied as transcription factor and growth factor. Secreted extracellularly by activated monocytes and macrophages or passively released by necrotic or damaged cells, extracellular HMGB1 is a potent mediator of inflammation. Extracellular HMGB1 has apparently contrasting biological actions: it sustains inflammation (with the possible establishment of autoimmunity or of self-maintaining tissue damage), but it also activates and recruits stem cells, boosting tissue repair. Here, we focus on the role of HMGB1 in physiological and pathological responses, the mechanisms by which it contributes to tissue repair and therapeutic strategies base on targeting HMGB1.
Collapse
Affiliation(s)
- Elia Ranzato
- Department of Sciences and Innovative Technology, (DiSIT), University of Piemonte Orientale "A. Avogadro", Viale Teresa Michel 11, Alessandria 15121, Italy.
| | - Simona Martinotti
- Department of Sciences and Innovative Technology, (DiSIT), University of Piemonte Orientale "A. Avogadro", Viale Teresa Michel 11, Alessandria 15121, Italy.
| | - Marco Pedrazzi
- Department of Experimental Medicine (DIMES)-Biochemistry Section, Center of Excellence for Biomedical Research (CEBR), University of Genoa, Viale Benedetto XV, Genoa 1-16132, Italy.
| | - Mauro Patrone
- Department of Sciences and Innovative Technology, (DiSIT), University of Piemonte Orientale "A. Avogadro", Viale Teresa Michel 11, Alessandria 15121, Italy.
| |
Collapse
|