1
|
Liu J, Fu Y, Jia W, Gao C, Tang H, Li H, Yang W. Multifunctional alginate-sinapic acid/arginine-strontium hydrogel for promoting diabetic wound healing. Int J Biol Macromol 2025; 308:142460. [PMID: 40154687 DOI: 10.1016/j.ijbiomac.2025.142460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/15/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
Sodium alginate (Alg), sinapic acid (SA), arginine (Arg), strontium carbonate (SrCO₃), and D-gluconic acid δ-lactone (GDL) were employed to construct an ionically crosslinked sodium alginate-sinapic acid/arginine‑strontium (ASASG) hydrogel. This study aims to evaluate its potential for enhancing diabetic wound repair through combined ionic crosslinking and bioactive component synergy. The hydrogel demonstrates exceptional swelling properties conducive to the absorption of wound exudate. Furthermore, it exhibits remarkable antibacterial and antioxidant activities. The ASASG hydrogel shows good biocompatibility, facilitates hemostasis and promotes the migration of NIH 3 T3 fibroblasts. Animal studies have revealed that ASASG hydrogel significantly enhances wound healing, accelerates re-epithelialization, and fosters collagen deposition in diabetic rats. Furthermore, ASASG hydrogels mitigated inflammatory cell infiltration by down-regulating IL-6 expression and concurrently up-regulating TGF-β expression. Notably, ASASG hydrogels also stimulate angiogenesis by enhancing the expressions of VEGF and bFGF. Therefore, the use of ASASG hydrogel in the treatment of diabetic wounds is a possible therapeutic approach.
Collapse
Affiliation(s)
- Jie Liu
- College of Pharmaceutical Science & Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, China State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China
| | - Ying Fu
- College of Pharmaceutical Science & Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, China State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China
| | - Wenlin Jia
- College of Pharmaceutical Science & Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, China State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China
| | - Chenrong Gao
- College of Pharmaceutical Science & Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, China State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China
| | - Hongbo Tang
- Department of Pharmacy, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, 100026, China.
| | - Haiying Li
- College of Pharmaceutical Science & Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, China State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China.
| | - Wenzhi Yang
- College of Pharmaceutical Science & Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, China State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Hebei University, Baoding 071002, China.
| |
Collapse
|
2
|
Astaneh ME, Hashemzadeh A, Fereydouni N. Recent advances in sodium alginate-based dressings for targeted drug delivery in the context of diabetic wound healing. J Mater Chem B 2024; 12:10163-10197. [PMID: 39283024 DOI: 10.1039/d4tb01049c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Diabetic wounds pose a significant challenge in healthcare due to impaired healing and increased risk of complications. In recent years, various drug delivery systems with stimuli-responsive features have been developed to address these issues. These systems enable precise dosage control and tailored drug release, promoting comprehensive tissue repair and regeneration. This review explores targeted therapeutic agents, such as carboxymethyl chitosan-alginate hydrogel formulations, nanofiber mats, and core-shell nanostructures, for diabetic wound healing. Additionally, the integration of nanotechnology and multifunctional biomimetic scaffolds shows promise in enhancing wound healing outcomes. Future research should focus on optimizing the design, materials, and printing parameters of 3D-bio-printed wound dressings, as well as exploring combined strategies involving the simultaneous release of antibiotics and nitric oxide for improved wound healing.
Collapse
Affiliation(s)
- Mohammad Ebrahim Astaneh
- Department of Anatomical Sciences, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Department of Tissue Engineering, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran.
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
| | - Alireza Hashemzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Narges Fereydouni
- Department of Tissue Engineering, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran.
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
3
|
Kim DY, Ko E, Ryu YH, Lee SJ, Jun YJ. Hyaluronic Acid Based Adipose Tissue-Derived Extracellular Matrix Scaffold in Wound Healing: Histological and Immunohistochemical Study. Tissue Eng Regen Med 2024; 21:829-842. [PMID: 38647955 PMCID: PMC11286915 DOI: 10.1007/s13770-024-00644-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND In this study, we explored the potential of human adipose tissue-derived extracellular matrix (adECM) sheets augmented with crosslinked hyaluronic acid (HA) as advanced wound dressings. We aimed to enhance healing efficacy while optimizing cost efficiency. METHODS The adECM was processed from healthy donor tissue and combined with crosslinked HA to form ECM-HA sheets (Scaffiller, Medikan, Korea). In vitro experiments involved seeding adipose-derived stem cells (ASCs) onto these sheets and assessing cell survival and cytokine production. In vivo testing utilized a rat wound model, comparing ECM-HA sheet with HA-based dressing and polyurethane foam dressing. Re-epithelialization and collagen deposition were examined through histopathological examinations, whereas immunohistochemistry was used to assess CD31, alpha smooth muscle actin (α-SMA), and Tenascin C expression as contributing factors to wound healing. RESULTS Results indicated that ECM-HA sheets were produced efficiently, with enhanced growth factor production and ASC survival observed in vitro. In vivo, ECM-HA sheets demonstrated accelerated wound healing, evidenced by improved epithelialization, thicker dermis, increased collagen deposition, and enhanced vascularity. Notably, they exhibited reduced myofibroblast activity and increased expression of Tenascin C, suggesting a favorable healing environment. CONCLUSION ECM-HA sheets offer a promising approach for wound management, combining the benefits of adECM and HA. They present improved stability and cost-effectiveness while promoting essential aspects of wound healing such as angiogenesis and collagen formation. This study underscores the therapeutic potential of ECM-HA sheets in clinical applications aimed at facilitating wound repair.
Collapse
Affiliation(s)
- Dong Yeon Kim
- Department of Plastic and Reconstructive Surgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eunjeong Ko
- Department of Plastic and Reconstructive Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-Gu, Seoul, 137-701, Republic of Korea
| | - Yeon Hee Ryu
- Department of Plastic and Reconstructive Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-Gu, Seoul, 137-701, Republic of Korea
| | - Su Jin Lee
- Department of Plastic and Reconstructive Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-Gu, Seoul, 137-701, Republic of Korea
| | - Young Joon Jun
- Department of Plastic and Reconstructive Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-Gu, Seoul, 137-701, Republic of Korea.
| |
Collapse
|
4
|
Esmaeilzadeh A, Yeganeh PM, Nazari M, Esmaeilzadeh K. Platelet-derived extracellular vesicles: a new-generation nanostructured tool for chronic wound healing. Nanomedicine (Lond) 2024; 19:915-941. [PMID: 38445377 DOI: 10.2217/nnm-2023-0344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024] Open
Abstract
Chronic nonhealing wounds pose a serious challenge to regaining skin function and integrity. Platelet-derived extracellular vesicles (PEVs) are nanostructured particles with the potential to promote wound healing since they can enhance neovascularization and cell migration and reduce inflammation and scarring. This work provides an innovative overview of the technical laboratory issues in PEV production, PEVs' role in chronic wound healing and the benefits and challenges in its clinical translation. The article also explores the challenges of proper sourcing, extraction techniques and storage conditions, and discusses the necessity of further evaluations and combinational therapeutics, including dressing biomaterials, M2-derived exosomes, mesenchymal stem cells-derived extracellular vesicles and microneedle technology, to boost their therapeutic efficacy as advanced strategies for wound healing.
Collapse
Affiliation(s)
- Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, 77978-45157, Iran
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, 77978-45157, Iran
| | | | - Mahdis Nazari
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, 77978-45157, Iran
| | - Kimia Esmaeilzadeh
- Department of Medical Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, 77978-45157, Iran
| |
Collapse
|
5
|
Tejedor S, Wågberg M, Correia C, Åvall K, Hölttä M, Hultin L, Lerche M, Davies N, Bergenhem N, Snijder A, Marlow T, Dönnes P, Fritsche-Danielson R, Synnergren J, Jennbacken K, Hansson K. The Combination of Vascular Endothelial Growth Factor A (VEGF-A) and Fibroblast Growth Factor 1 (FGF1) Modified mRNA Improves Wound Healing in Diabetic Mice: An Ex Vivo and In Vivo Investigation. Cells 2024; 13:414. [PMID: 38474378 PMCID: PMC10930761 DOI: 10.3390/cells13050414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Diabetic foot ulcers (DFU) pose a significant health risk in diabetic patients, with insufficient revascularization during wound healing being the primary cause. This study aimed to assess microvessel sprouting and wound healing capabilities using vascular endothelial growth factor (VEGF-A) and a modified fibroblast growth factor (FGF1). METHODS An ex vivo aortic ring rodent model and an in vivo wound healing model in diabetic mice were employed to evaluate the microvessel sprouting and wound healing capabilities of VEGF-A and a modified FGF1 both as monotherapies and in combination. RESULTS The combination of VEGF-A and FGF1 demonstrated increased vascular sprouting in the ex vivo mouse aortic ring model, and topical administration of a combination of VEGF-A and FGF1 mRNAs formulated in lipid nanoparticles (LNPs) in mouse skin wounds promoted faster wound closure and increased neovascularization seven days post-surgical wound creation. RNA-sequencing analysis of skin samples at day three post-wound creation revealed a strong transcriptional response of the wound healing process, with the combined treatment showing significant enrichment of genes linked to skin growth. CONCLUSION f-LNPs encapsulating VEGF-A and FGF1 mRNAs present a promising approach to improving the scarring process in DFU.
Collapse
Affiliation(s)
- Sandra Tejedor
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
- Systems Biology Research Center, School of Bioscience, University of Skövde, 541 28 Skövde, Sweden; (P.D.); (J.S.)
| | - Maria Wågberg
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
| | - Cláudia Correia
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
| | - Karin Åvall
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
| | - Mikko Hölttä
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
| | - Leif Hultin
- Imaging and Data Analytics, Clinical and Pharmacological Safety Science, BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden;
| | - Michael Lerche
- Advanced Drug Delivery, Pharmaceutical Science, BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden; (M.L.); (N.D.)
| | - Nigel Davies
- Advanced Drug Delivery, Pharmaceutical Science, BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden; (M.L.); (N.D.)
| | - Nils Bergenhem
- Alliance Management, Business Development and Licensing, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA 02451, USA
| | - Arjan Snijder
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden; (A.S.)
| | - Tom Marlow
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden; (A.S.)
| | - Pierre Dönnes
- Systems Biology Research Center, School of Bioscience, University of Skövde, 541 28 Skövde, Sweden; (P.D.); (J.S.)
- SciCross AB, 541 35 Skövde, Sweden
| | - Regina Fritsche-Danielson
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
| | - Jane Synnergren
- Systems Biology Research Center, School of Bioscience, University of Skövde, 541 28 Skövde, Sweden; (P.D.); (J.S.)
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Karin Jennbacken
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
| | - Kenny Hansson
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
| |
Collapse
|
6
|
Zuo L, Zhu S, Gu S, Xu X. Anti-scarring effects of conbercept on human Tenon's fibroblasts: comparisons with bevacizumab. BMC Ophthalmol 2023; 23:183. [PMID: 37101202 PMCID: PMC10131424 DOI: 10.1186/s12886-023-02914-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 04/08/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Safely inhibiting the formation of scar in the glaucoma filtration surgery (GFS) has always been an issue for clinical glaucoma doctors. Anti-vascular endothelial growth factor (VEGF) agents can reduce angiogenesis, and anti-placental growth factor (PIGF) agents can affect reactive gliosis. However, the effect of conbercept, which can bind to both VEGF and PIGF, on human Tenon's fibroblasts (HTFs) is unknown. METHODS HTFs were cultured in vitro and treated with conbercept or bevacizumab (BVZ). No drug was added to the control group. The effects of drugs on cell proliferation were assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, and the collagen type I alpha1(Col1A1) mRNA expression level was measured using quantitative polymerase chain reaction (qPCR). HTF cell migration after drug interventions was evaluated using the scratch wound assay along with the measurement of the expression levels of VEGF and PIGF in human umbilical vein endothelial cells (HUVECs) using enzyme-linked immunosorbent assay, as well as the detection of the VEGF(R) mRNA expression level in HTFs using qPCR. RESULTS After the addition of conbercept (0.01, 0.1, and 1 mg/mL) to the cultured HTFs or HUVECs, no significant cytotoxicity was observed compared with the control group, while the cytotoxicity of 2.5 mg/mL BVZ on HTFs was obvious. Conbercept significantly inhibited HTF cell migration and Col1A1 mRNA expression level in HTFs. It was superior to BVZ in inhibiting HTF migration. After the intervention with conbercept, the expression level of PIGF and VEGF in HUVECs significantly decreased; and the inhibitory effect of conbercept on the expression level of VEGF in HUVECs was weaker than that of BVZ. Conbercept was more advantageous than BVZ in inhibiting the expression level of VEGFR-1 mRNA in HTFs. However, its effect in terms of inhibiting the expression level of VEGFR-2 mRNA in HTFs was weaker than that of BVZ. CONCLUSION The results suggested the low cytotoxicity and significant anti-scarring effect of conbercept in HTF with significant anti-PIGF and inferior anti-VEGF effects compared with BVZ, thus providing a better understanding of the role of conbercept in the GFS wound healing process.
Collapse
Affiliation(s)
- Lei Zuo
- Department of Ophthalmology, Shanghai Fourth People's Hospital, Tongji University School of Medicine, No. 1279, Sanmen road, Shanghai, 200434, China
| | - Shaopin Zhu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 85 / 86, Wujin road, Shanghai, 200080, China
| | - Shengjie Gu
- Department of Ophthalmology, Shanghai Fourth People's Hospital, Tongji University School of Medicine, No. 1279, Sanmen road, Shanghai, 200434, China
| | - Xun Xu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 85 / 86, Wujin road, Shanghai, 200080, China.
| |
Collapse
|
7
|
Highly concentrated trehalose induces prohealing senescence-like state in fibroblasts via CDKN1A/p21. Commun Biol 2023; 6:13. [PMID: 36609486 PMCID: PMC9822918 DOI: 10.1038/s42003-022-04408-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 12/23/2022] [Indexed: 01/08/2023] Open
Abstract
Trehalose is the nonreducing disaccharide of glucose, evolutionarily conserved in invertebrates. The living skin equivalent (LSE) is an organotypic coculture containing keratinocytes cultivated on fibroblast-populated dermal substitutes. We demonstrated that human primary fibroblasts treated with highly concentrated trehalose promote significantly extensive spread of the epidermal layer of LSE without any deleterious effects. The RNA-seq analysis of trehalose-treated 2D and 3D fibroblasts at early time points revealed the involvement of the CDKN1A pathway, the knockdown of which significantly suppressed the upregulation of DPT, ANGPT2, VEGFA, EREG, and FGF2. The trehalose-treated fibroblasts were positive for senescence-associated β-galactosidase. Finally, transplantation of the dermal substitute with trehalose-treated fibroblasts accelerated wound closure and increased capillary formation significantly in the experimental mouse wounds in vivo, which was canceled by the CDKN1A knockdown. These data indicate that high-concentration trehalose can induce the senescence-like state in fibroblasts via CDKN1A/p21, which may be therapeutically useful for optimal wound repair.
Collapse
|
8
|
Porcine placental extract increase the cellular NAD levels in human epidermal keratinocytes. Sci Rep 2022; 12:19040. [PMID: 36352014 PMCID: PMC9646745 DOI: 10.1038/s41598-022-23446-9] [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: 06/02/2022] [Accepted: 10/31/2022] [Indexed: 11/11/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is an essential cofactor for numerous enzymes involved in energy metabolism. Because decreasing NAD levels is a common hallmark of the aging process in various tissues and organs, maintaining NAD levels has recently been of interest for the prevention of aging and age-related diseases. Although placental extract (PE) are known to possess several anti-aging effects, the NAD-boosting activity of PE remains unknown. In this study, we found that porcine PE (PPE) significantly increased intracellular NAD levels in normal human epidermal keratinocytes (NHEKs). PPE also attenuated the NAD depletion induced by FK866, an inhibitor of nicotinamide phosphoribosyltransferase (NAMPT). Interestingly, only the fraction containing nicotinamide mononucleotide (NMN), nicotinamide riboside (NR), and nicotinamide (NAM) restored NAD content in NHEKs in the absence of NAMPT activity. These results suggest that PPE increases intracellular NAD by providing NAD precursors such as NMN, NR, and NAM. Finally, we showed that the application of PPE to the stratum corneum of the reconstructed human epidermis significantly ameliorated FK866-induced NAD depletion, suggesting that topical PPE may be helpful for increasing skin NAD levels. This is the first study to report the novel biological activity of PE as an NAD booster in human epidermal cells.
Collapse
|
9
|
Dardenne C, Salon M, Authier H, Meunier E, AlaEddine M, Bernad J, Bouschbacher M, Lefèvre L, Pipy B, Coste A. Topical Aspirin Administration Improves Cutaneous Wound Healing in Diabetic Mice Through a Phenotypic Switch of Wound Macrophages Toward an Anti-inflammatory and Proresolutive Profile Characterized by LXA4 Release. Diabetes 2022; 71:2181-2196. [PMID: 35796692 DOI: 10.2337/db20-1245] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 06/29/2022] [Indexed: 11/13/2022]
Abstract
Patients with diabetes present a persistent inflammatory process, leading to impaired wound healing. Since nonhealing diabetic wound management shows limited results, the introduction of advanced therapies targeting and correcting the inflammatory status of macrophages in chronic wounds could be an effective therapeutic strategy to stop the sustained inflammation and to return to a healing state. In an excisional skin injury in a diet-induced diabetic murine model, we demonstrate that topical administration of low-dose aspirin (36 μg/wound/day) improves cutaneous wound healing by increasing wound closure through the promotion of the inflammation resolution program of macrophages. This treatment increased efferocytosis of wound macrophages from aspirin-treated diabetic mice compared with untreated diabetic mice. We also show that aspirin treatment of high-fat-fed mice oriented the phenotype of wound macrophages toward an anti-inflammatory and proresolutive profile characterized by a decrease of LTB4 production. The use of diabetic mice deficient for 5-LOX or 12/15-LOX demonstrated that these two enzymes of acid arachidonic metabolism are essential for the beneficial effect of aspirin on wound healing. Thus, aspirin treatment modified the balance between pro- and anti-inflammatory eicosanoids by promoting the synthesis of proresolving LXA4 through 5-LOX, LTA4, 12/15-LOX signaling. In conclusion, the restoration of an anti-inflammatory and proresolutive phenotype of wound macrophages by the topical administration of low-dose aspirin represents a promising therapeutic approach in chronic wounds.
Collapse
Affiliation(s)
- Christophe Dardenne
- UMR 152 PHARMA-DEV, Université de Toulouse, and Institut de Recherche pour le Développement, Université Paul Sabatier, Toulouse, France
| | - Marie Salon
- UMR 152 PHARMA-DEV, Université de Toulouse, and Institut de Recherche pour le Développement, Université Paul Sabatier, Toulouse, France
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, Université Paul Sabatier, Toulouse, France
| | - Hélène Authier
- UMR 152 PHARMA-DEV, Université de Toulouse, and Institut de Recherche pour le Développement, Université Paul Sabatier, Toulouse, France
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, Université Paul Sabatier, Toulouse, France
| | - Etienne Meunier
- UMR 5089, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
| | - Mohamad AlaEddine
- UMR 152 PHARMA-DEV, Université de Toulouse, and Institut de Recherche pour le Développement, Université Paul Sabatier, Toulouse, France
| | - José Bernad
- UMR 152 PHARMA-DEV, Université de Toulouse, and Institut de Recherche pour le Développement, Université Paul Sabatier, Toulouse, France
| | | | - Lise Lefèvre
- UMR 152 PHARMA-DEV, Université de Toulouse, and Institut de Recherche pour le Développement, Université Paul Sabatier, Toulouse, France
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, Université Paul Sabatier, Toulouse, France
| | - Bernard Pipy
- UMR 152 PHARMA-DEV, Université de Toulouse, and Institut de Recherche pour le Développement, Université Paul Sabatier, Toulouse, France
| | - Agnès Coste
- UMR 152 PHARMA-DEV, Université de Toulouse, and Institut de Recherche pour le Développement, Université Paul Sabatier, Toulouse, France
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
10
|
Awasthi A, Vishwas S, Gulati M, Corrie L, Kaur J, Khursheed R, Alam A, Alkhayl FF, Khan FR, Nagarethinam S, Kumar R, Arya K, Kumar B, Chellappan DK, Gupta G, Dua K, Singh SK. Expanding arsenal against diabetic wounds using nanomedicines and nanomaterials: Success so far and bottlenecks. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103534] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
11
|
Zhu L, Qian J, Jiang Y, Yang T, Duan Q, Xiao X. PlGF Reduction Compromises Angiogenesis in Diabetic Foot Disease Through Macrophages. Front Immunol 2021; 12:736153. [PMID: 34659227 PMCID: PMC8511710 DOI: 10.3389/fimmu.2021.736153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 09/13/2021] [Indexed: 12/19/2022] Open
Abstract
Diabetic foot disease (DFD) is a common and serious complication for diabetes and is characterized with impaired angiogenesis. In addition to the well-defined role of vascular endothelial growth factor (VEGF) -A and its defect in the pathogenesis of DFD, another VEGF family member, placental growth factor (PlGF), was also recently found to alter expression pattern in the DFD patients with undetermined mechanisms. This question was thus addressed in the current study. We detected attenuated PlGF upregulation in a mouse DFD model. In addition, the major cell types at the wound to express the unique PlGF receptor, VEGF receptor 1 (VEGFR1), were macrophages and endothelial cells. To assess how PlGF regulates DFD-associated angiogenesis, we injected recombinant PlGF and depleted VEGF1R specifically in macrophages by local injection of an adeno-associated virus (AAV) carrying siRNA for VEGFR1 under a macrophage-specific CD68 promoter. We found that the angiogenesis and recovery of the DFD were both improved by PlGF injection. The PlGF-induced improvement in angiogenesis and the recovery of skin injury were largely attenuated by macrophage-specific depletion of VEGF1R, likely resulting from reduced macrophage number and reduced M2 polarization. Together, our data suggest that reduced PlGF compromises angiogenesis in DFD at least partially through macrophages.
Collapse
Affiliation(s)
- Lingyan Zhu
- Department of Endocrinology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Endocrinology, The Peoples Hospital of Yudu County, Ganzhou, China
| | - Jieqi Qian
- Department of Surgery, Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yinan Jiang
- Department of Surgery, Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Tianlun Yang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China
| | - Qiong Duan
- Department of Cardiology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiangwei Xiao
- Department of Surgery, Children’s Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
12
|
Schönborn M, Łączak P, Pasieka P, Borys S, Płotek A, Maga P. Pro- and Anti-Angiogenic Factors: Their Relevance in Diabetic Foot Syndrome-A Review. Angiology 2021; 73:299-311. [PMID: 34541892 DOI: 10.1177/00033197211042684] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Peripheral arterial disease can involve tissue loss in up to 50% of patients with diabetic foot syndrome (DFS). Consequently, revascularization of narrowed or occluded arteries is one of the most common forms of comprehensive treatment. However, technically successful angioplasty does not always result in the healing of ulcers. The pathomechanism of this phenomenon is still not fully understood, but inadequate angiogenesis in tissue repair may play an essential role. Changes in pro- and anti-angiogenic factors among patients with DFS are not always clear and conclusive. In particular, some studies underline the role of decreased concentration of pro-angiogenic factors and higher levels of anti-angiogenic mediators. Nevertheless, there are still controversial issues, including the paradox of impaired wound healing despite high concentrations of some pro-angiogenic factors, dynamics of their expression during the healing process, and their mutual relationships. Exploring this process among diabetic patients may provide new insight into well-known methods of treatment and show their real benefits and chances for improving outcomes.
Collapse
Affiliation(s)
- Martyna Schönborn
- Department of Angiology, Faculty of Medicine, 162261Jagiellonian University Medical College, Krakow, Poland.,Doctoral School of Medical and Health Sciences, 162261Jagiellonian University, Krakow, Poland
| | - Patrycja Łączak
- Department of Angiology, Faculty of Medicine, 162261Jagiellonian University Medical College, Krakow, Poland
| | - Paweł Pasieka
- Department of Angiology, Faculty of Medicine, 162261Jagiellonian University Medical College, Krakow, Poland
| | - Sebastian Borys
- Department of Metabolic Diseases, Faculty of Medicine, 162261Jagiellonian University Medical College, Krakow, Poland
| | - Anna Płotek
- Department of Angiology, Faculty of Medicine, 162261Jagiellonian University Medical College, Krakow, Poland
| | - Paweł Maga
- Department of Angiology, Faculty of Medicine, 162261Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
13
|
Colotti G, Failla CM, Lacal PM, Ungarelli M, Ruffini F, Di Micco P, Orecchia A, Morea V. Neuropilin-1 is required for endothelial cell adhesion to soluble vascular endothelial growth factor receptor 1. FEBS J 2021; 289:183-198. [PMID: 34252269 PMCID: PMC9290910 DOI: 10.1111/febs.16119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 05/27/2021] [Accepted: 07/12/2021] [Indexed: 12/22/2022]
Abstract
Neuropilin‐1 (NRP‐1) is a semaphorin receptor involved in neuron guidance, and a co‐receptor for selected isoforms of the vascular endothelial growth factor (VEGF) family. NRP‐1 binding to several VEGF‐A isoforms promotes growth factor interaction with VEGF receptor (VEGFR)‐2, increasing receptor phosphorylation. Additionally, NRP‐1 directly interacts with VEGFR‐1, but this interaction competes with NRP‐1 binding to VEGF‐A165 and does not enhance VEGFR‐1 activation. In this work, we investigated in detail the role of NRP‐1 interaction with the soluble isoform of VEGFR‐1 (sVEGFR‐1) in angiogenesis. sVEGFR‐1 acts both as a decoy receptor for VEGFs and as an extracellular matrix protein directly binding to α5β1 integrin on endothelial cells. By combining cell adhesion assays and surface plasmon resonance experiments on purified proteins, we found that sVEGFR‐1/NRP‐1 interaction is required both for α5β1 integrin binding to sVEGFR‐1 and for endothelial cell adhesion to a sVEGFR‐1‐containing matrix. We also found that a previously reported anti‐angiogenic peptide (Flt2‐11), which maps in the second VEGFR‐1 Ig‐like domain, specifically binds NRP‐1 and inhibits NRP‐1/sVEGFR‐1 interaction, a process that likely contributes to its anti‐angiogenic activity. In view of potential translational applications, we developed a five‐residue‐long peptide, derived from Flt2‐11, which has the same ability as the parent Flt2‐11 peptide to inhibit cell adhesion to, and migration towards, sVEGFR‐1. Therefore, the Flt2‐5 peptide represents a potential anti‐angiogenic compound per se, as well as an attractive lead for the development of novel angiogenesis inhibitors acting with a different mechanism with respect to currently used therapeutics, which interfere with VEGF‐A165 binding.
Collapse
Affiliation(s)
- Gianni Colotti
- Institute of Molecular Biology and Pathology (IBPM) of the National Research Council (CNR), Rome, Italy
| | | | | | | | | | - Patrizio Di Micco
- Department of Biochemical Sciences 'A. Rossi Fanelli', Sapienza' University of Rome, Italy
| | - Angela Orecchia
- Laboratory of Molecular and Cell Biology, IDI-IRCCS, Rome, Italy
| | - Veronica Morea
- Institute of Molecular Biology and Pathology (IBPM) of the National Research Council (CNR), Rome, Italy
| |
Collapse
|
14
|
Pawar KB, Desai S, Bhonde RR, Bhole RP, Deshmukh AA. Wound with Diabetes: Present Scenario and Future. Curr Diabetes Rev 2021; 17:136-142. [PMID: 32619172 DOI: 10.2174/1573399816666200703180137] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/12/2020] [Accepted: 06/18/2020] [Indexed: 11/22/2022]
Abstract
Diabetes is a chronic metabolic disorder of the endocrine system characterized by an increase in blood glucose level. Several factors, such as pancreatic damage, oxidative stress, infection, genetic factor, obesity, liver dysfunction, play a vital role in the pathogenesis of diabetes, which further leads to serious diabetic complications. The diabetic wound is one such complication where the wound formation occurs, especially due to pressure and its healing process is disrupted due to factors, such as hyperglycemia, neuropathy, nephropathy, peripheral vascular disease, reduction of blood flow, atherosclerosis, impaired fibroblast. The process of wound healing is delayed due to different abnormalities like alteration in nitric oxide level, increase in aldose reductase, sorbitol, and fructose. Therefore, diabetic wound requires more time to heal as compared to the normal wound. Healing time is delayed in diabetic wound due to many factors, such as stress, decreased oxygenation supply, infection, decreased blood flow, impaired proliferation and migration rate, impaired growth factor production, impaired keratinocytes proliferation and migration, and altered vascular endothelial mediators. The current treatment for diabetic wounds includes wound patches, oxygenation therapy, hydrogel patches, gene therapy, laser therapy, and stem cell therapy. Medications with phytoconstituents are also one way to manage the diabetic wound, but it is not more effective for quick healing. The objective of this review is to understand the potential of various management options which are available for diabetic wound, with a special focus on biological cells.
Collapse
Affiliation(s)
- Kuldeep B Pawar
- Department of Pharmacology, Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pune, India
| | - Shivani Desai
- Department of Pharmacology, Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pune, India
| | | | - Ritesh P Bhole
- Department of Pharmaceutical Chemistry, Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pune, India
| | - Atul A Deshmukh
- Department of Pharmacology, Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pune, India
| |
Collapse
|
15
|
Scull G, Brown AC. Development of novel microenvironments for promoting enhanced wound healing. CURRENT TISSUE MICROENVIRONMENT REPORTS 2020; 1:73-87. [PMID: 33748773 PMCID: PMC7968354 DOI: 10.1007/s43152-020-00009-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Nonhealing wounds are a significant issue facing the healthcare industry. Materials that modulate the wound microenvironment have the potential to improve healing outcomes. RECENT FINDINGS A variety of acellular and cellular scaffolds have been developed for regulating the wound microenvironment, including materials for controlled release of antimicrobials and growth factors, materials with inherent immunomodulative properties, and novel colloidal-based scaffolds. Scaffold construction methods include electrospinning, 3D printing, decellularization of extracellular matrix, or a combination of techniques. Material application methods include layering or injecting at the wound site. SUMMARY Though these techniques show promise for repairing wounds, all material strategies thus far struggle to induce regeneration of features such as sweat glands and hair follicles. Nonetheless, innovative technologies currently in the research phase may facilitate future attainment of these features. Novel methods and materials are constantly arising for the development of microenvironments for enhanced wound healing.
Collapse
Affiliation(s)
- Grant Scull
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Raleigh, NC 27695
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27695
| | - Ashley C. Brown
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Raleigh, NC 27695
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27695
| |
Collapse
|
16
|
Advances in the Research of Bioinks Based on Natural Collagen, Polysaccharide and Their Derivatives for Skin 3D Bioprinting. Polymers (Basel) 2020; 12:polym12061237. [PMID: 32485901 PMCID: PMC7362214 DOI: 10.3390/polym12061237] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/23/2020] [Accepted: 02/25/2020] [Indexed: 12/22/2022] Open
Abstract
The skin plays an important role in protecting the human body, and wound healing must be set in motion immediately following injury or trauma to restore the normal structure and function of skin. The extracellular matrix component of the skin mainly consists of collagen, glycosaminoglycan (GAG), elastin and hyaluronic acid (HA). Recently, natural collagen, polysaccharide and their derivatives such as collagen, gelatin, alginate, chitosan and pectin have been selected as the matrix materials of bioink to construct a functional artificial skin due to their biocompatible and biodegradable properties by 3D bioprinting, which is a revolutionary technology with the potential to transform both research and medical therapeutics. In this review, we outline the current skin bioprinting technologies and the bioink components for skin bioprinting. We also summarize the bioink products practiced in research recently and current challenges to guide future research to develop in a promising direction. While there are challenges regarding currently available skin bioprinting, addressing these issues will facilitate the rapid advancement of 3D skin bioprinting and its ability to mimic the native anatomy and physiology of skin and surrounding tissues in the future.
Collapse
|
17
|
Huynh P, Phie J, Krishna SM, Golledge J. Systematic review and meta-analysis of mouse models of diabetes-associated ulcers. BMJ Open Diabetes Res Care 2020; 8:e000982. [PMID: 32467222 PMCID: PMC7259859 DOI: 10.1136/bmjdrc-2019-000982] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/29/2020] [Accepted: 04/18/2020] [Indexed: 12/14/2022] Open
Abstract
Mouse models are frequently used to study diabetes-associated ulcers, however, whether these models accurately simulate impaired wound healing has not been thoroughly investigated. This systematic review aimed to determine whether wound healing is impaired in mouse models of diabetes and assess the quality of the past research. A systematic literature search was performed of publicly available databases to identify original articles examining wound healing in mouse models of diabetes. A meta-analysis was performed to examine the effect of diabetes on wound healing rate using random effect models. A meta-regression was performed to examine the effect of diabetes duration on wound healing impairment. The quality of the included studies was also assessed using two newly developed tools. 77 studies using eight different models of diabetes within 678 non-diabetic and 720 diabetic mice were included. Meta-analysis showed that wound healing was impaired in all eight models. Meta-regression suggested that longer duration of diabetes prior to wound induction was correlated with greater degree of wound healing impairment. Pairwise comparisons suggested that non-obese diabetic mice exhibited more severe wound healing impairment compared with db/db mice, streptozotocin-induced diabetic mice or high-fat fed mice at an intermediate stage of wound healing (p<0.01). Quality assessment suggested that the prior research frequently lacked incorporation of key clinically relevant characteristics. This systematic review suggested that impaired wound healing can be simulated in many different mouse models of diabetes but these require further refinement to become more clinically relevant.
Collapse
Affiliation(s)
- Pacific Huynh
- Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia
| | - James Phie
- Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia
| | - Smriti Murali Krishna
- Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia
- Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, Queensland, Australia
| |
Collapse
|
18
|
Saddala MS, Lennikov A, Huang H. Placental growth factor regulates the pentose phosphate pathway and antioxidant defense systems in human retinal endothelial cells. J Proteomics 2020; 217:103682. [PMID: 32058040 DOI: 10.1016/j.jprot.2020.103682] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 02/08/2020] [Indexed: 01/16/2023]
Abstract
The molecular mechanisms whereby placental growth factor (PlGF) mediates its effects in nonproliferative diabetic retinopathy (DR) are unknown. To better understand the role of PlGF in DR, we used tandem mass tags (TMT)-labeled quantitative proteomics to human retinal endothelial cells (HRECs), treated anti-PlGF antibody, and PBS as a control. Functional annotation and pathway enrichments were performed, which suggested that the differentially expressed proteins (DEPs) were involved in key metabolic processes, protein binding, and membrane, pentose phosphate pathway (PPP) and adherens junction. We conducted integrated gene profiles of our previously published transcriptomic data to the TMT-labeled proteomics data. The results showed the sixty genes were found to be changed at the proteome level. The functional annotation conducted for the sixty proteins suggested that 58.3% of proteins were involved in PPP, 25% of proteins were in interleukin-12 singling and 16.7% of proteins were involved in glycolysis and gluconeogenesis pathway. Mass spectrometry results were validated by transendothelial electrical resistance measurement by an electrical cell-impedance sensing (ECIS) and western blot analysis of VE-cadherin, G6PD. These findings suggest that the PPP proteins and antioxidants may act as a downstream target of PlGF and may play a decisive role in HREC biological functions in DR. SIGNIFICANCE: PlGF (Placental growth factor) is known to play a pivotal role in pathological angiogenesis and inflammation by stimulating endothelial cell migration and by recruiting pericytes and inflammatory cells such as microglia and macrophages. Despite the well-defined pathophysiological roles of PlGF, the underlying molecular and cellular mechanisms are not completely understood, especially the exact relationships between biochemical events and molecular pathways regulated by PlGF, whose inhibition exhibits a protective role in DR. This study provides new insights into protein expression patterns and enables the identification of many attractive candidates for investigation of PPP pathway role in the activation of the antioxidant defense system in DR. Our findings suggest that the PPP proteins and antioxidants (PRDX6, HMOX1, NQO1 and YES1) may act as downstream targets of PlGF and may play a decisive role in HREC biological functions in DR.
Collapse
Affiliation(s)
| | - Anton Lennikov
- Mason Eye Institute, University of Missouri, Columbia, MO, United States
| | - Hu Huang
- Mason Eye Institute, University of Missouri, Columbia, MO, United States.
| |
Collapse
|
19
|
Ishida Y, Kuninaka Y, Nosaka M, Furuta M, Kimura A, Taruya A, Yamamoto H, Shimada E, Akiyama M, Mukaida N, Kondo T. CCL2-Mediated Reversal of Impaired Skin Wound Healing in Diabetic Mice by Normalization of Neovascularization and Collagen Accumulation. J Invest Dermatol 2019; 139:2517-2527.e5. [PMID: 31247201 DOI: 10.1016/j.jid.2019.05.022] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 12/14/2022]
Abstract
Patients with diabetes frequently present with complications such as impaired skin wound healing. Skin wound sites display a markedly enhanced expression of CCL2, a potent macrophage chemoattractant, together with macrophage infiltration during the early inflammatory phase in skin wound healing of healthy individuals, but the association of CCL2 with delayed skin wound healing in patients with diabetes remains elusive. In this study, we showed that, compared with control mice, mice with streptozotocin-induced diabetes displayed impaired healing after excisional skin injury, with decreased neovascularization, CCL2 expression, and macrophage infiltration. Compromised skin wound healing in mice with diabetes was reversed by the administration of topical CCL2 immediately after the injury, as evidenced by normalization of wound closure rates, neovascularization, collagen accumulation, and infiltration of macrophages expressing vascular endothelial growth factor, a potent angiogenic factor, and transforming growth factor-β. CCL2 treatment further increased the accumulation of endothelial progenitor cells at the wound sites of mice with diabetes and eventually accelerated neovascularization. Thus, the topical application of CCL2 can be an effective therapeutic option for the treatment of patients with diabetes with defective wound repair, promoting neovascularization and collagen accumulation at skin wound sites.
Collapse
Affiliation(s)
- Yuko Ishida
- Department of Forensic Medicine, Wakayama Medical University, Kimiidera, Wakayama, Japan
| | - Yumi Kuninaka
- Department of Forensic Medicine, Wakayama Medical University, Kimiidera, Wakayama, Japan
| | - Mizuho Nosaka
- Department of Forensic Medicine, Wakayama Medical University, Kimiidera, Wakayama, Japan
| | - Machi Furuta
- Clinical Laboratory Medicine, Wakayama Medical University, Kimiidera, Wakayama, Japan
| | - Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, Kimiidera, Wakayama, Japan
| | - Akira Taruya
- Cardiovascular Medicine, Wakayama Medical University, Kimiidera, Wakayama, Japan
| | - Hiroki Yamamoto
- Department of Forensic Medicine, Wakayama Medical University, Kimiidera, Wakayama, Japan
| | - Emi Shimada
- Department of Forensic Medicine, Wakayama Medical University, Kimiidera, Wakayama, Japan
| | - Mariko Akiyama
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, Kimiidera, Wakayama, Japan.
| |
Collapse
|
20
|
Huang H, Lennikov A, Saddala MS, Gozal D, Grab DJ, Khalyfa A, Fan L. Placental growth factor negatively regulates retinal endothelial cell barrier function through suppression of glucose-6-phosphate dehydrogenase and antioxidant defense systems. FASEB J 2019; 33:13695-13709. [PMID: 31585507 DOI: 10.1096/fj.201901353r] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We report that placental growth factor (PlGF) negatively affects the endothelial cell (EC) barrier function through a novel regulatory mechanism. The PlGF mAb promotes (but recombinant protein disrupts) EC barrier function, thus affecting the barrier-forming protein levels, membrane distribution, and EC monolayer impedance by the electrical cell-impedance sensing system, Western blot, and immunofluorescence staining. RNA sequencing-based transcriptome analysis identified the up-regulation of the pentose phosphate pathway (PPP) and the antioxidant defense protein by PlGF blockade. The PlGF and PlGF/VEGF dimers (but not VEGF-A) down-regulated the protein expression of glucose-6-phosphate dehydrogenase (G6PD) and peroxiredoxin (PRDX). G6PD inhibition and gene silencing (small interfering RNA) abolished the beneficial effects of PlGF inhibition on EC barrier function and PRDX3/6 protein expression. VEGF receptor (VEGFR)1 or VEGFR2 blockade prevented the inhibitory effect of PlGF on G6PD protein expression and EC barrier function. The PRDX6 played dual roles in EC barrier function through glutathione peroxidase and phospholipase A2 activity. In sum, PlGF negatively regulates EC barrier function through the activation of VEGFR1 and VEGFR2 and the suppression of the G6PD/PPP and the antioxidant pathways.-Huang, H., Lennikov, A., Saddala, M. S., Gozal, D., Grab, D. J., Khalyfa, A., Fan, L. Placental growth factor negatively regulates endothelial cell barrier function through suppression of glucose-6-phosphate dehydrogenase and antioxidant defense systems.
Collapse
Affiliation(s)
- Hu Huang
- Department of Ophthalmology, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Anton Lennikov
- Department of Ophthalmology, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Madhu Sudhana Saddala
- Department of Ophthalmology, University of Missouri-Columbia, Columbia, Missouri, USA
| | - David Gozal
- Child Health Research Institute, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Dennis J Grab
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA.,Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Abdelnaby Khalyfa
- Child Health Research Institute, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Lijuan Fan
- Department of Ophthalmology, University of Missouri-Columbia, Columbia, Missouri, USA
| |
Collapse
|
21
|
Silva AT, Rouf F, Semola OA, Payton ME, Lovern PC. Placental growth factor levels in quadriceps muscle are reduced by a Western diet in association with advanced glycation end products. Am J Physiol Heart Circ Physiol 2019; 317:H851-H866. [PMID: 31397166 DOI: 10.1152/ajpheart.00511.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In peripheral artery disease (PAD), atherosclerotic occlusion chronically impairs limb blood flow. Arteriogenesis (collateral artery remodeling) is a vital adaptive response to PAD that protects tissue from ischemia. People with type II diabetes have a high risk of developing PAD and would benefit from arteriogenesis. However, arteriogenesis is suppressed in people with diabetes by a multifaceted mechanism which remains incompletely defined. Upregulation of placental growth factor (PLGF) is a key early step in arteriogenesis. Therefore, we hypothesized that metabolic dysfunction would impair PLGF expression in skeletal muscle. We tested this hypothesis in C57BL/6J and ApoE-/- mice of both sexes fed a Western diet (WD) for 24 wk. We first assessed baseline levels of PLGF, vascular endothelial growth factor (VEGF-A), and VEGF receptor 1 (VEGFR1) protein in hindlimb skeletal muscle. Only PLGF was consistently decreased by the WD. We next investigated the effect of 24 wk of the WD on the response of PLGF, VEGF-A, VEGFR1, and monocyte chemoattractant protein-1 (MCP-1) to the physiological stimulus of vascular occlusion. Hindlimb ischemia was induced in mice by gradual femoral artery occlusion using an ameroid constrictor. Growth factor levels were measured 3-28 days postsurgery. In C57BL/6J mice, the WD decreased and delayed upregulation of PLGF and abolished upregulation of VEGF-A and VEGFR1 but had no effect on MCP-1. In ApoE-/- mice fed either diet, all factors tested failed to respond to occlusion. Metabolic phenotyping of mice and in vitro studies suggest that an advanced glycation end product/TNFα-mediated mechanism could contribute to the effects observed in vivo.NEW & NOTEWORTHY In this study, we tested the effect of a Western diet on expression of the arteriogenic growth factor placental growth factor (PLGF) in mouse skeletal muscle. We provide the first demonstration that a Western diet interferes with both baseline expression and hindlimb ischemia-induced upregulation of PLGF. We further identify a potential role for advanced glycation end product/TNFα signaling as a negative regulator of PLGF. These studies provide insight into one possible mechanism by which type II diabetes may limit collateral growth.
Collapse
Affiliation(s)
- Asitha T Silva
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Farzana Rouf
- Department of Mechanical and Aerospace Engineering, Oklahoma State University, Stillwater, Oklahoma
| | - Oluwayemisi A Semola
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Mark E Payton
- Department of Statistics, Oklahoma State University, Stillwater, Oklahoma
| | - Pamela C Lovern
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma
| |
Collapse
|
22
|
Kwon TR, Han SW, Kim JH, Lee BC, Kim JM, Hong JY, Kim BJ. Polydeoxyribonucleotides Improve Diabetic Wound Healing in Mouse Animal Model for Experimental Validation. Ann Dermatol 2019; 31:403-413. [PMID: 33911618 PMCID: PMC7992745 DOI: 10.5021/ad.2019.31.4.403] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 01/02/2019] [Accepted: 02/12/2019] [Indexed: 12/13/2022] Open
Abstract
Background Wound healing mechanisms is believed to have effects similar to wound healing disorders in diabetic patients, including abnormal inflammatory cells, angiogenesis disorders, and reduced collagen synthesis. Therefore, reestablishment of structural and promoted angiogenesis could be beneficial to promote wound healing process. Objective Therefore, we investigated whether the polydeoxyribonucleotide (PDRN) that was self-production in Korea, could be useful as an intradermal injection for promoting wound healing. Also, we validate for wound healing effect of PDRN using healing-impaired (db/db) mice. Methods In this study, we confirmed the effects of PDRN by creating wound models in in vitro and in vivo model. Using an in vitro wound healing assay, we observed that PDRN stimulated closure of wounded monolayers of human fibroblast cells. PDRN (8.25 mg/ml) or phosphate-buffered saline (0.9% NaCl) was injected once daily into the dermis adjacent to the wound for 12 days after skin injury. Results Time course observations revealed that mice treated with PDRN showed accelerated wound closure and epidermal and dermal regeneration, enhanced angiogenesis. The wound area and depth decreased at 3, 6, 9, and 12 days after skin injury. Histological evaluation showed an increase of vascular endothelial growth factor, CD31, and collagen fibers in the PDRN group compared with the control group, indicating that PDRN was effective in the treatment of delayed wound healing caused by diabetes. Conclusion This study suggests that our PDRN has a wound healing effect in transgenic animal models with cells and diabetes through angiogenesis.
Collapse
Affiliation(s)
- Tae-Rin Kwon
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Sung Won Han
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea.,Department of Medicine, Graduate School, Chung-Ang University, Seoul, Korea
| | - Jong Hwan Kim
- Department of Medicine, Graduate School, Chung-Ang University, Seoul, Korea
| | - Byung Chul Lee
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea.,Department of Medicine, Graduate School, Chung-Ang University, Seoul, Korea
| | - Jae Min Kim
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Ji Yeon Hong
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Beom Joon Kim
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea.,Department of Medicine, Graduate School, Chung-Ang University, Seoul, Korea
| |
Collapse
|
23
|
Veith AP, Henderson K, Spencer A, Sligar AD, Baker AB. Therapeutic strategies for enhancing angiogenesis in wound healing. Adv Drug Deliv Rev 2019; 146:97-125. [PMID: 30267742 DOI: 10.1016/j.addr.2018.09.010] [Citation(s) in RCA: 581] [Impact Index Per Article: 96.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 09/15/2018] [Accepted: 09/24/2018] [Indexed: 12/19/2022]
Abstract
The enhancement of wound healing has been a goal of medical practitioners for thousands of years. The development of chronic, non-healing wounds is a persistent medical problem that drives patient morbidity and increases healthcare costs. A key aspect of many non-healing wounds is the reduced presence of vessel growth through the process of angiogenesis. This review surveys the creation of new treatments for healing cutaneous wounds through therapeutic angiogenesis. In particular, we discuss the challenges and advancement that have been made in delivering biologic, pharmaceutical and cell-based therapies as enhancers of wound vascularity and healing.
Collapse
|
24
|
Huppertz B. Biology of preeclampsia: Combined actions of angiogenic factors, their receptors and placental proteins. Biochim Biophys Acta Mol Basis Dis 2018; 1866:165349. [PMID: 30553017 DOI: 10.1016/j.bbadis.2018.11.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/07/2018] [Accepted: 11/22/2018] [Indexed: 12/13/2022]
Abstract
Although massive efforts have been undertaken to elucidate the etiology of the pregnancy syndrome preeclampsia, its developmental origin remains a mystery. Most efforts of the last decade have focused on biomarkers to predict and/or diagnose preeclampsia, including the anti-angiogenic factor sFlt-1 (soluble fms-like tyrosin kinase-1), the angiogenic factor PGF (placental growth factor) and PP13 (placental protein 13). The origins of these marker proteins are still under debate, and so far their actions have only been describe separate from each other. This study will focus on the origins and actions of all three markers during pregnancy and outside pregnancy and will describe a scenario where all three markers act synergistically to rescue the mother from the deleterious effects of the debris that is released from the placenta during preeclampsia. This more holistic approach may open new avenues to think about maternal-fetal interactions and putative therapies.
Collapse
Affiliation(s)
- Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria.
| |
Collapse
|
25
|
Heo JH, Heo Y, Lee HJ, Kim M, Shin HY. Topical anti-inflammatory and anti-oxidative effects of porcine placenta extracts on 2,4-dinitrochlorobenzene-induced contact dermatitis. Altern Ther Health Med 2018; 18:331. [PMID: 30541534 PMCID: PMC6291973 DOI: 10.1186/s12906-018-2396-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 11/29/2018] [Indexed: 11/10/2022]
Abstract
Background The placenta is a reservoir enriched with growth factors, hormones, cytokines and minerals. While several beneficial effects of placenta extracts on wound healing, anti-aging and anti-inflammatory responses have been reported, relatively limited mechanistic exploration has been conducted to date. Here, we provide compelling evidence of anti-inflammatory and anti-oxidative activities of porcine placenta extracts (PPE) against contact dermatitis in vivo. Methods A contact dermatitis mouse model was established by sensitizing the dorsal skin of BALB/c mice using the contact allergen, 2,4-dinitrochlorobenzene (DNCB), and molecular consequences of topical application of PPE were investigated. PPEs were pre-sterilized via γ-irradiation, which is a milder but more effective way of sterilizing biomolecules relative to the conventional autoclaving method. Results DNCB-induced skin lesions displayed clear contact dermatitis-like symptoms and topical application of PPE dramatically alleviated both local and systemic inflammatory responses. Inflammatory epidermal thickening was completely abrogated and allergen-specific serum IgE levels significantly reduced in the presence of PPE. Moreover, anti-oxidative activities of PPE were observed both in vitro and in vivo, which may lead to attenuation of inflammatory responses. Prolonged treatment with PPE strongly inhibited production of DNCB-induced reactive oxygen species (ROS) and subsequently prevented oxidative degradation of hyaluronic acid (HA), which triggers innate inflammatory responses. Conclusion Our findings supply valuable insights into the mechanisms underlying the anti-inflammatory effects of PPE and provide a functional basis for the clinical application of PPE in inflammatory diseases. Electronic supplementary material The online version of this article (10.1186/s12906-018-2396-1) contains supplementary material, which is available to authorized users.
Collapse
|
26
|
Huppertz B. An updated view on the origin and use of angiogenic biomarkers for preeclampsia. Expert Rev Mol Diagn 2018; 18:1053-1061. [PMID: 30413130 DOI: 10.1080/14737159.2018.1546579] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Introduction: The last decade has seen massive efforts towards the identification and the potential use of predictive biomarkers for the pregnancy pathology preeclampsia. The angiogenic factors sFlt-1 and placental growth factor (PGF) have been in focus and have been massively supported. Areas covered: This review describes preeclampsia and intra-uterine growth restriction (IUGR), focusing on sFlt-1 and PGF, their sources during and outside pregnancy and the application of these markers in diseases outside pregnancy. Finally, the specificity of the angiogenic markers for preeclampsia is discussed. Expert commentary: The admixture of the two independent syndromes preeclampsia and IUGR has not helped in identifying the etiologies of either. Rather, it has made the search for new markers and pathways much more complicated as has the constriction on the angiogenic markers. The current markers sFlt-1 and PGF have a clear value once an adverse outcome is diagnosed but are not specific for preeclampsia. Also, they are mostly derived from the maternal vascular system rather than the placenta and are already in use as markers outside pregnancy. A new holistic approach using disease maps and interoperable workflows based on topic-related big data will help in broadening our understanding of the etiology of preeclampsia and hence, develop new markers and therapies.
Collapse
Affiliation(s)
- Berthold Huppertz
- a Division of Cell Biology, Histology and Embryology , Gottfried Schatz Research Center, Medical University of Graz , Graz , Austria
| |
Collapse
|
27
|
The role of placental growth factor (PlGF) and its receptor system in retinal vascular diseases. Prog Retin Eye Res 2018; 69:116-136. [PMID: 30385175 DOI: 10.1016/j.preteyeres.2018.10.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/23/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022]
Abstract
Placental growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family. Upon binding to VEGF- and neuropilin-receptor sub-types, PlGF modulates a range of neural, glial and vascular cell responses that are distinct from VEGF-A. As PlGF expression is selectively associated with pathological angiogenesis and inflammation, its blockade does not affect the healthy vasculature. PlGF actions have been extensively described in tumor biology but more recently there has been accumulating preclinical evidence that indicates that this growth factor could have an important role in retinal diseases. High levels of PlGF have been found in aqueous humor, vitreous and/or retina of patients exhibiting retinopathies, especially those with diabetic retinopathy (DR) and neovascular age-related macular degeneration (nvAMD). Expression of this growth factor seems to correlate closely with many of the key pathogenic features of early and late retinopathy in preclinical models. For example, studies using genetic modification and/or pharmacological treatment to block PlGF in the laser-induced choroidal neovascularization (CNV) model, oxygen-induced retinopathy model, as well as various murine diabetic models, have shown that PlGF deletion or inhibition can reduce neovascularization, retinal leakage, inflammation and gliosis, without affecting vascular development or inducing neuronal degeneration. Moreover, an inhibitory effect of PlGF blockade on retinal scarring in the mouse CNV model has also been recently demonstrated and was found to be unique for PlGF inhibition, as compared to various VEGF inhibition strategies. Together, these preclinical results suggest that anti-PlGF therapy might have advantages over anti-VEGF treatment, and that it may have clinical applications as a standalone treatment or in combination with anti-VEGF. Additional clinical studies are clearly needed to further elucidate the role of PlGF and its potential as a therapeutic target in ocular diseases.
Collapse
|
28
|
Wise LM, Stuart GS, Real NC, Fleming SB, Mercer AA. VEGF Receptor-2 Activation Mediated by VEGF-E Limits Scar Tissue Formation Following Cutaneous Injury. Adv Wound Care (New Rochelle) 2018; 7:283-297. [PMID: 30087804 DOI: 10.1089/wound.2016.0721] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/27/2017] [Indexed: 02/06/2023] Open
Abstract
Objective: Vascular endothelial growth factor (VEGF) family members are critical regulators of tissue repair and depending on their distinct pattern of receptor specificity can also promote inflammation and scarring. This study utilized a receptor-selective VEGF to examine the role of VEGF receptor (VEGFR)-2 in scar tissue (ST) formation. Approach: Cutaneous skin wounds were created in mice using a 4 mm biopsy punch and then treated until closure with purified VEGF-E derived from orf virus stain NZ-2. Tissue samples were harvested to measure gene expression using quantitative PCR and to observe ST formation through histological examination and changes in cell populations by immunofluorescence. Results: VEGFR-2-activation with VEGF-E increased expression of anti-inflammatory cytokine interleukin (IL)-10 and reduced macrophage infiltration and myofibroblast differentiation in wounded skin compared with controls. VEGF-E treatment also increased microvascular density and improved pericyte coverage of blood vessels in the healing wounds. The ST that formed following treatment with VEGF-E was reduced in size and showed improved collagen structure. Innovation: The role of VEGFR-2 activation in wound epithelialization and angiogenesis is well established; but its contribution to ST formation is unclear. This study tests the effect of a selective VEGFR-2 activation on ST formation following cutaneous wounding in a murine model. Conclusion: VEGFR-2 stimulation can enhance the quality of skin repair, at least, in part, through the induction of IL-10 expression and dampening of wound inflammation and fibrosis. Therapies that selectively activate VEGFR-2 may therefore be beneficial to treat impaired healing or to prevent excess scarring.
Collapse
Affiliation(s)
- Lyn M. Wise
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Gabriella S. Stuart
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Nicola C. Real
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Stephen B. Fleming
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Andrew A. Mercer
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
29
|
Chen Z, Fu S, Wu Z, Chen J, Huang Y, Wang Y, Fu M. Relationship between plasma angiogenic growth factors and diabetic foot ulcers. Clin Chim Acta 2018; 482:95-100. [DOI: 10.1016/j.cca.2018.03.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/17/2018] [Accepted: 03/27/2018] [Indexed: 12/19/2022]
|
30
|
Accelerated growth of hemangioblastoma in pregnancy: the role of proangiogenic factors and upregulation of hypoxia-inducible factor (HIF) in a non-oxygen-dependent pathway. Neurosurg Rev 2017; 42:209-226. [DOI: 10.1007/s10143-017-0910-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/16/2017] [Accepted: 09/19/2017] [Indexed: 12/28/2022]
|
31
|
Van Bergen T, Hu TT, Etienne I, Reyns GE, Moons L, Feyen JHM. Neutralization of placental growth factor as a novel treatment option in diabetic retinopathy. Exp Eye Res 2017; 165:136-150. [PMID: 28965804 DOI: 10.1016/j.exer.2017.09.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 09/08/2017] [Accepted: 09/27/2017] [Indexed: 12/11/2022]
Abstract
The current standard of care in clinical practice for diabetic retinopathy (DR), anti-vascular endothelial growth factor (VEGF) therapy, has shown a significant improvement in visual acuity. However, treatment response can be variable and might be associated with potential side effects. This study was designed to investigate inhibition of placental growth factor (PlGF) as a possible alternative therapy for DR. The effect of the anti-PlGF antibody (PL5D11D4) was preclinically evaluated in various animal models by investigating different DR hallmarks, including inflammation, neurodegeneration, vascular leakage and fibrosis. The in vivo efficacy was tested in diabetic streptozotocin (STZ) and Akimba models and in the laser induced choroidal neovascularization (CNV) mouse model. Intravitreal (IVT) administration of the anti-PlGF antibody was compared to anti-VEGFR-2 antibody (DC101), anti-VEGF antibody (B20), VEGF-Trap (aflibercept) and triamcinolone acetonide (TAAC). Vascular leakage was investigated in the mouse STZ model by fluorescein isothiocyanate labeled bovine serum albumin (FITC-BSA) perfusion and in the Akimba model by fluorescein angiography (FA). Repeated IVT administration of the anti-PlGF antibody reduced vascular leakage, which was comparable to a single administration of VEGFR-2 inhibition in the mouse STZ model. PL5D11D4 treatment did not alter retinal ganglion cell (RGC) density, as demonstrated by Brn3a staining, whereas DC101 significantly reduced RGC number with 20%. Immunohistological stainings were performed to investigate inflammation (CD45, F4/80) and fibrosis (collagen type 1a). In the CNV model, IVT injection(s) of PL5D11D4 dose-dependently reduced inflammation and fibrosis, as compared to PBS treatment. Equimolar single administration of the anti-PlGF antibody and aflibercept (21 nM) and TAAC decreased leukocyte and macrophage infiltration with 50%, whereas DC101 and B20 (21 nM) had no effect on the inflammatory response. Similar results were observed in the mouse STZ model on the number of microglia and macrophages in the retina. Repeated administration of PL5D11D4 (21 nM) and TAAC similarly reduced fibrosis, while no effect was observed after equimolar DC101, B20 nor aflibercept administration (21 nM). In summary, the anti-PlGF antibody showed comparable efficacy as well-characterized VEGF-inhibitor on the process of vascular leakage, but differentiates itself by also reducing inflammation and fibrosis, without triggering a neurodegenerative response.
Collapse
Affiliation(s)
- Tine Van Bergen
- ThromboGenics NV, Gaston Geenslaan 1, 3001 Heverlee, Belgium.
| | - Tjing-Tjing Hu
- ThromboGenics NV, Gaston Geenslaan 1, 3001 Heverlee, Belgium.
| | | | - Geert E Reyns
- ThromboGenics NV, Gaston Geenslaan 1, 3001 Heverlee, Belgium.
| | - Lieve Moons
- Department of Biology, Zoological Institute, KU Leuven, Leuven, Belgium.
| | - Jean H M Feyen
- ThromboGenics NV, Gaston Geenslaan 1, 3001 Heverlee, Belgium.
| |
Collapse
|
32
|
Nejabati HR, Latifi Z, Ghasemnejad T, Fattahi A, Nouri M. Placental growth factor (PlGF) as an angiogenic/inflammatory switcher: lesson from early pregnancy losses. Gynecol Endocrinol 2017; 33:668-674. [PMID: 28447504 DOI: 10.1080/09513590.2017.1318375] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Placental growth factor (PlGF) is an angiogenic factor which belongs to vascular endothelial growth factor (VEGF) family. In addition to the angiogenic function of PlGF, in some conditions such as preeclampsia and early pregnancy losses, it can induce inflammatory reactions which could be accompanied with reduced angiogenesis. Hence, it is crucial to investigate inflammatory and angiogenic switching states and understand underlying mechanisms. PlGF is expressed in endometrium, placenta and trophoblast cells and is involved in maturation of uterine NK cells. Up-regulation of PlGF directs VEGF to VEGFR-2 and reinforces angiogenesis. However, when VEGF/VEGFR-2 signaling pathway is impaired, PlGF may shift to severe inflammation and cause tissue damages which could lead to early pregnancy losses. Downregulation of PlGF has also been reported in pregnancy complications. In this review, we discussed the role of PlGF in embryo implantation failure and early pregnancy loss and also possible mechanisms regarding the role of PlGF in angiogenic/inflammatory switching in early pregnancy losses. Furthermore, we summarized the effects of various compounds on PlGF expression and briefly talked about its therapeutic potential that may be an opportunity for prevention of pregnancy loss.
Collapse
Affiliation(s)
- Hamid Reza Nejabati
- a Women's Reproductive Health Research Center
- b Department of Clinical Biochemistry , Faculty of Medicine , and
| | - Zeinab Latifi
- b Department of Clinical Biochemistry , Faculty of Medicine , and
| | | | - Amir Fattahi
- a Women's Reproductive Health Research Center
- c Department of Reproductive Biology , Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Mohammad Nouri
- c Department of Reproductive Biology , Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
33
|
Zhang Y, Liu NM, Wang Y, Youn JY, Cai H. Endothelial cell calpain as a critical modulator of angiogenesis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1326-1335. [PMID: 28366876 DOI: 10.1016/j.bbadis.2017.03.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 03/04/2017] [Accepted: 03/28/2017] [Indexed: 12/13/2022]
Abstract
Calpains are a family of calcium-dependent non-lysosomal cysteine proteases. In particular, calpains residing in the endothelial cells play important roles in angiogenesis. It has been shown that calpain activity can be increased in endothelial cells by growth factors, primarily vascular endothelial growth factor (VEGF). VEGF/VEGFR2 induces calpain 2 dependent activation of PI3K/AMPK/Akt/eNOS pathway, and consequent nitric oxide production and physiological angiogenesis. Under pathological conditions such as tumor angiogenesis, endothelial calpains can be activated by hypoxia. This review focuses on the molecular regulatory mechanisms of calpain activation, and the newly identified mechanistic roles and downstream signaling events of calpains in physiological angiogenesis, and in the conditions of pathological tumor angiogenesis and diabetic wound healing, as well as retinopathy and atherosclerosis that are also associated with an increase in calpain activity. Further discussed include the differential strategies of modulating angiogenesis through manipulating calpain expression/activity in different pathological settings. Targeted limitation of angiogenesis in cancer and targeted promotion of angiogenesis in diabetic wound healing via modulations of calpains and calpain-dependent signaling mechanisms are of significant translational potential. Emerging strategies of tissue-specific targeting, environment-dependent targeting, and genome-targeted editing may turn out to be effective regimens for targeted manipulation of angiogenesis through calpain pathways, for differential treatments including both attenuation of tumor angiogenesis and potentiation of diabetic angiogenesis.
Collapse
Affiliation(s)
- Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Norika Mengchia Liu
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Yongchen Wang
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Ji Youn Youn
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA; Division of Cardiology, Department Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California Los Angeles (UCLA), CA 90095, USA.
| |
Collapse
|
34
|
Photobiomodulation laser and pulsed electrical field increase the viability of the musculocutaneous flap in diabetic rats. Lasers Med Sci 2017; 32:641-648. [DOI: 10.1007/s10103-017-2160-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/25/2017] [Indexed: 01/09/2023]
|
35
|
Activated Transcription Factor 3 in Association with Histone Deacetylase 6 Negatively Regulates MicroRNA 199a2 Transcription by Chromatin Remodeling and Reduces Endothelin-1 Expression. Mol Cell Biol 2016; 36:2838-2854. [PMID: 27573019 DOI: 10.1128/mcb.00345-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/25/2016] [Indexed: 01/18/2023] Open
Abstract
Previous studies showed that high levels of placenta growth factor (PlGF) correlated with increased plasma levels of endothelin-1 (ET-1), a potent vasoconstrictor, in sickle cell disease (SCD). PlGF-mediated transcription of the ET-1 gene occurs by activation of hypoxia inducible factor 1α (HIF-1α) and posttranscriptionally by microRNA 199a2 (miR-199a2), which targets the 3' untranslated region (UTR) of HIF-1α mRNA. However, relatively less is known about how PlGF represses the expression of miR-199a2 located in the DNM3 opposite strand (DNM3os) transcription unit. Here, we show that PlGF induces the expression of activated transcription factor 3 (ATF3), which, in association with accessory proteins (c-Jun dimerization protein 2 [JDP2], ATF2, and histone deacetylase 6 [HDAC6]), as determined by proteomic analysis, binds to the DNM3os promoter. Furthermore, we show that association of HDAC6 with ATF3 at its binding site in this promoter was correlated with repression of miR-199a2 transcription, as shown by DNM3os transcription reporter and chromatin immunoprecipitation (ChIP) assays. Tubacin, an inhibitor of HDAC6, antagonized PlGF-mediated repression of DNM3os/pre-miR-199a2 transcription with a concomitant reduction in ET-1 levels in cultured endothelial cells. Analysis of lung tissues from Berkeley sickle (BK-SS) mice showed increased levels of ATF3 and increased expression of ET-1. Delivery of tubacin to BK-SS mice significantly attenuated plasma ET-1 and PlGF levels. Our studies demonstrated that ATF3 in conjunction with HDAC6 acts as a transcriptional repressor of the DNM3os/miR-199a2 locus.
Collapse
|
36
|
Vascular Endothelial Growth Factor Receptor Type 1 Signaling Prevents Delayed Wound Healing in Diabetes by Attenuating the Production of IL-1β by Recruited Macrophages. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1481-98. [DOI: 10.1016/j.ajpath.2016.02.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 01/01/2016] [Accepted: 02/01/2016] [Indexed: 12/17/2022]
|
37
|
Avitabile S, Odorisio T, Madonna S, Eyerich S, Guerra L, Eyerich K, Zambruno G, Cavani A, Cianfarani F. Interleukin-22 Promotes Wound Repair in Diabetes by Improving Keratinocyte Pro-Healing Functions. J Invest Dermatol 2015; 135:2862-2870. [PMID: 26168231 DOI: 10.1038/jid.2015.278] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/08/2015] [Accepted: 06/18/2015] [Indexed: 11/09/2022]
Abstract
Impaired re-epithelialization, imbalanced expression of cytokines and growth factors, and vascular disease contribute to healing impairment in diabetes. IL-22, a pro-inflammatory cytokine mediating a cross-talk between immune system and epithelial cells, has been shown to have a role in repair processes. In this study we aimed to investigate IL-22 regenerative potential in the poor healing context of diabetic wounds. By using streptozotocin-induced diabetic mice, we demonstrated that IL-22 wound treatment significantly accelerated the healing process, by promoting re-epithelialization, granulation tissue formation, and vascularization. Improved re-epithelialization was associated with increased keratinocyte proliferation and signal transducer and activator of transcription 3 (STAT3) activation. We showed that endogenous IL-22 content was reduced at both mRNA and protein level during the inflammatory phase of diabetic wounds, with fewer IL-22-positive cells infiltrating the granulation tissue. We demonstrated that IL-22 treatment promoted proliferation and injury repair of hyperglycemic keratinocytes and induced activation of STAT3 and extracellular signal-regulated kinase transduction pathways in keratinocytes grown in hyperglycemic condition or isolated from diabetic patients. Finally, we demonstrated that IL-22 treatment was able to inhibit diabetic keratinocyte differentiation while promoting vascular endothelial growth factor release. Our data indicate a pro-healing role of IL-22 in diabetic wounds, suggesting a therapeutic potential for this cytokine in diabetic ulcer management.
Collapse
Affiliation(s)
- Simona Avitabile
- Laboratory of Experimental Immunology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Teresa Odorisio
- Laboratory of Biochemistry, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Stefania Madonna
- Laboratory of Experimental Immunology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Stefanie Eyerich
- ZAUM - Center of Allergy and Environment, Technische Universität and Helmholtz Center, Munich, Germany
| | - Liliana Guerra
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Kilian Eyerich
- Department of Dermatology and Allergy, Technische Universität, Munich, Germany
| | - Giovanna Zambruno
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Andrea Cavani
- Laboratory of Experimental Immunology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy.
| | - Francesca Cianfarani
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy.
| |
Collapse
|
38
|
Sharma K, Bhattacharyya D. Immunoglobulin isotype isolated from human placental extract does not interfere in complement-mediated bacterial opsonization within the wound milieu. FEBS Open Bio 2015; 5:369-77. [PMID: 25984442 PMCID: PMC4431336 DOI: 10.1016/j.fob.2015.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 04/08/2015] [Accepted: 04/10/2015] [Indexed: 11/29/2022] Open
Abstract
Human placental extract has many applications as a wound healer. Immunoglobulin G is a key glycoprotein present in human placental extract. Placental IgG (25.2 ± 3.97 μg/ml) did not exert anti-complementary effects.
The wound healing potency of an aqueous extract of placenta can be evaluated through the presence of numerous regulatory components. The presence of glycans was detected by thin layer chromatography and fluorophore-assisted carbohydrate electrophoresis. Mass spectrometric analysis revealed the existence of multiple fragments of immunoglobulin G (IgG). IgG was present in the extract at a concentration of 25.2 ± 3.97 μg/ml. IgG possesses anti-complementary activity by diverting the complement activation from target surface. Thus, effect of placental IgG on complement–bacteria interaction was investigated through classical and alternative pathway and the preparation was ascertained to be safe with respect to their interference in the process of bacterial opsonization.
Collapse
Key Words
- ANTS, 8-aminonaphthalene-1,3,6-trisulfonate
- ATP, adenosine triphosphate
- BCIP, 5-Bromo 4-Chloro 3′ indolylphosphate
- BHI, Brain–Heart Infusion
- BSA, bovine serum albumin
- CNBr, cyanogens bromide
- Complement
- EDTA, ethylenediamine tetra acetic acid
- EGTA, ethylene glycol tetra acetic acid
- G6PDH, glucose-6-phosphate dehydrogenase
- Human placental extract
- IgG, immunoglobulin G
- Immunoglobulin
- NAD+, nicotinamide adenine dinucleotide
- NBT, nitro-blue tetrazolium chloride
- PNGase F, peptide N-glycosidase F
- Pseudomonas aeruginosa wound physiology
Collapse
Affiliation(s)
- Kanika Sharma
- Division of Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C.Mullick Road, Jadavpur, Kolkata 7000032, India
| | - Debasish Bhattacharyya
- Division of Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C.Mullick Road, Jadavpur, Kolkata 7000032, India
| |
Collapse
|
39
|
Does Topical Application of Placental Extract Gel on Postoperative Fibrotomy Wound Improve Mouth Opening and Wound Healing in Patients With Oral Submucous Fibrosis? J Oral Maxillofac Surg 2015; 73:1439.e1-10. [PMID: 25891654 DOI: 10.1016/j.joms.2015.03.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 03/03/2015] [Accepted: 03/05/2015] [Indexed: 10/23/2022]
Abstract
PURPOSE Placental extract has been used as a therapeutic agent with application in various fields of medicine. Placental extract is well known for its effects on wound healing with anti-inflammatory, antiplatelet, and angiogenic effects and is also a biogenic modulator. The present study evaluated the effect of placental extract on wound healing, mouth opening, and postoperative patient discomfort in patients with oral submucous fibrosis treated with fibrotomy with buccal fat pad coverage and coronoidectomy. MATERIALS AND METHODS Ten subjects with oral submucous fibrosis who presented with mouth opening less than 20 mm were enrolled in the present prospective randomized controlled trial to assess the effects of placental extract on the fibrotomy wound covered with a pedicled buccal pad fat (5 patients allocated to the study group, group S and 5 to the control group, group C). The following criteria were used to analyze the postoperative effect of placental extract on fibrotomy wounds compared with that of the controls: subjective assessment of the wound, postoperative discomfort, and postoperative mouth opening assessed at 1, 2, and 4 weeks postoperatively. RESULTS The average difference in the preoperative and fourth week postoperative mouth opening for group C was 13.8 ± 2.68 mm and was 21.20 ± 2.77 mm in group S. The median calculated for group C was a 15.0-mm increase in mouth opening and was 20.0 mm in group S. CONCLUSION The results obtained with topical application of placental extract on fibrotomy wound healing and postoperative mouth opening were superior to those of the control group in whom placental extract was not used.
Collapse
|
40
|
Erythropoietin-mediated expression of placenta growth factor is regulated via activation of hypoxia-inducible factor-1α and post-transcriptionally by miR-214 in sickle cell disease. Biochem J 2015; 468:409-23. [PMID: 25876995 DOI: 10.1042/bj20141138] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 04/16/2015] [Indexed: 12/31/2022]
Abstract
Placental growth factor (PlGF) plays an important role in various pathological conditions and diseases such as inflammation, cancer, atherosclerosis and sickle cell disease (SCD). Abnormally high PlGF levels in SCD patients are associated with increased inflammation and pulmonary hypertension (PHT) and reactive airway disease; however, the transcriptional and post-transcriptional mechanisms regulating PlGF expression are not well defined. Herein, we show that treatment of human erythroid cells and colony forming units with erythropoietin (EPO) increased PlGF expression. Our studies showed EPO-mediated activation of HIF-1α led to subsequent binding of HIF-1α to hypoxia response elements (HREs) within the PlGF promoter, as demonstrated by luciferase transcription reporter assays and ChIP analysis of the endogenous gene. Additionally, we showed miR-214 post-transcriptionally regulated the expression of PlGF as demonstrated by luciferase reporter assays using wild-type (wt) and mutant PlGF-3'-UTR constructs. Furthermore, synthesis of miR-214, located in an intron of DNM3 (dynamin 3), was transcriptionally regulated by transcription factors, peroxisome proliferator-activated receptor-α (PPARα) and hypoxia-inducible factor-1α (HIF-1α). These results were corroborated in vivo wherein plasma from SCD patients and lung tissues from sickle mice showed an inverse correlation between PlGF and miR-214 levels. Finally, we observed that miR-214 expression could be induced by fenofibrate, a Food and Drug Administration (FDA) approved PPARα agonist, thus revealing a potential therapeutic approach for reduction in PlGF levels by increasing miR-214 transcription. This strategy has potential clinical implications for several pathological conditions including SCD.
Collapse
|
41
|
Kristen AV, Rinn J, Hegenbart U, Lindenmaier D, Merkle C, Röcken C, Hardt S, Giannitsis E, Katus HA. Improvement of risk assessment in systemic light-chain amyloidosis using human placental growth factor. Clin Res Cardiol 2014; 104:250-7. [PMID: 25331161 DOI: 10.1007/s00392-014-0779-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/15/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND Vascular amyloid deposition is common in light-chain amyloidosis resulting in endothelial dysfunction. Human placental growth factor (PlGF), a member of the vascular endothelial growth factor family was found to be altered in diverse pathological conditions, e.g. endothelial dysfunction. This study evaluated the clinical role of PlGF in light-chain amyloidosis. METHODS PlGF (cobas-PlGF, Roche Diagnostics, Mannheim, Germany) was analyzed in 125 consecutive patients with AL and correlated with diverse clinical parameters including mortality. RESULTS Kidney (n = 76) and heart (n = 57) were predominantly affected by amyloid deposition. Median PlGF was 26.3 (21.1-42.1) ng/L, NT-proBNP 3649 (1124-8581) pg/mL, and hs-TnT 42 (21-107) ng/L. PlGF increased with number of organs involved and with deterioration of renal function. A significant correlation of PlGF with hs-TnT (ρ = 0.306; p = 0.0007) and NT-proBNP (ρ = 0.315; p = 0.0006) was observed, but no correlation was observed with clinical, echocardiography, and electrocardiography parameters of cardiac involvement. In this cohort 1-year all-cause mortality was 19.2 %. The best cutoff discriminating survivors and non-survivors was 28.44 ng/L (sensitivity 66.7 %; specificity 78.1 %). A three-step risk model including hs-TnT and NT-proBNP revealed a better discrimination if patients at intermediary risk were additionally stratified by PlGF. Net reclassification index was 37.2 % (p = 0.002). Multivariate analysis revealed PlGF, difference of involved and uninvolved light chain, number of organs involved and risk class according to troponin T and NT-proBNP as independent predictors of mortality. CONCLUSION Plasma PlGF values in AL are invariably associated with the number of involved organs, but not with clinical, echocardiography, and electrocardiography parameters of cardiac involvement. PlGF provide useful information for risk stratification of patients at intermediary risk according to hs-TnT and NT-proBNP.
Collapse
Affiliation(s)
- Arnt V Kristen
- Department of Cardiology, Angiology, and Respiratory Medicine, Heidelberg University, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany,
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Gacche RN, Meshram RJ. Angiogenic factors as potential drug target: Efficacy and limitations of anti-angiogenic therapy. Biochim Biophys Acta Rev Cancer 2014; 1846:161-79. [DOI: 10.1016/j.bbcan.2014.05.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/05/2014] [Accepted: 05/07/2014] [Indexed: 12/17/2022]
|
43
|
Choi HY, Kim SW, Kim B, Lee HN, Kim SJ, Song M, Kim S, Kim J, Kim YB, Kim JH, Cho SG. Alpha-fetoprotein, identified as a novel marker for the antioxidant effect of placental extract, exhibits synergistic antioxidant activity in the presence of estradiol. PLoS One 2014; 9:e99421. [PMID: 24922551 PMCID: PMC4055621 DOI: 10.1371/journal.pone.0099421] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 05/14/2014] [Indexed: 11/19/2022] Open
Abstract
Placenta, as a reservoir of nutrients, has been widely used in medical and cosmetic materials. Here, we focused on the antioxidant properties of placental extract and attempted to isolate and identify the main antioxidant factors. Porcine placental extracts were prepared through homogenization or acid hydrolysis, and their antioxidant activity was investigated in the human keratinocyte HaCaT cell line. Treatment with homogenized placental extract (H-PE) increased the cell viability of H2O2-treated HaCaT cells more than two-fold. H-PE treatment suppressed H2O2-induced apoptotic and necrotic cell death and decreased intracellular ROS levels in H2O2-treated HaCaT cells. The antioxidant factors in H-PE were found to be thermo-unstable and were thus expected to include proteins. The candidate antioxidant proteins were fractionated with cation-exchange, anion-exchange, and size-exclusion chromatography, and the antioxidant properties of the chromatographic fractions were investigated. We obtained specific antioxidant fractions that suppressed ROS generation and ROS-induced DNA strand breaks. From silver staining and MALDI-TOF analyses, alpha-fetoprotein (AFP) precursor was identified as a main marker for the antioxidant effect of H-PE. Purified AFP or ectopically expressed AFP exhibited synergistic antioxidant activity in the presence of estradiol. Taken together, our data suggest that AFP, a serum glycoprotein produced at high levels during fetal development, is a novel marker protein for the antioxidant effect of the placenta that exhibits synergistic antioxidant activity in the presence of estradiol.
Collapse
Affiliation(s)
- Hye Yeon Choi
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Republic of Korea
| | - Seung Woo Kim
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Republic of Korea
| | - BongWoo Kim
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Republic of Korea
| | - Hae Na Lee
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Republic of Korea
| | - Su-Jeong Kim
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Republic of Korea
| | - Minjung Song
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Republic of Korea
| | - Sol Kim
- Department of Life Science, Sogang University, Seoul, Republic of Korea
| | - Jungho Kim
- Department of Life Science, Sogang University, Seoul, Republic of Korea
| | - Young Bong Kim
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Republic of Korea
| | - Jin-Hoi Kim
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Republic of Korea
| | - Ssang-Goo Cho
- Department of Animal Biotechnology, Animal Resources Research Center, and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
44
|
Di Zenzo G, El Hachem M, Diociaiuti A, Boldrini R, Calabresi V, Cianfarani F, Fortugno P, Piccinni E, Zambruno G, Castiglia D. A truncating mutation in the laminin-332α chain highlights the role of the LG45 proteolytic domain in regulating keratinocyte adhesion and migration. Br J Dermatol 2014; 170:1056-64. [PMID: 24387836 DOI: 10.1111/bjd.12816] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2014] [Indexed: 01/21/2023]
Abstract
BACKGROUND Altered function of laminin-332 (α3β3γ2) consequent to mutations in the LAMA3, LAMB3 and LAMC2 genes causes junctional epidermolysis bullosa non-Herlitz (JEB-nH). JEB-nH patients suffer from skin blistering and have an increased risk of developing aggressive skin carcinomas in adulthood. Laminin-332 is proteolytically processed and its extracellular mature form lacks the α3 chain C-terminal globules 4 and 5 (LG45). The LG45 tandem has cell adhesion and protumorigenic properties. However, mutations that affect this domain are very rare and their functional effects in patients have not been explored to date. OBJECTIVE To characterize molecularly an adult patient with JEB-nH and altered laminin-332 expression presenting multiple skin carcinomas, and to analyse LG45-mediated biological functions using keratinocytes from the patient. METHODS A mutational search in laminin-332 genes was performed by hetero-duplex analysis. LAMA3 mRNA and laminin-332 protein levels in patient keratinocytes were investigated by real-time reverse transcriptase polymerase chain reaction and radioimmunoprecipitation assay, respectively. Keratinocyte migration was examined by scratch and Boyden chamber assays. RESULTS We identified a homozygous LAMA3 mutation, p.Leu1648TrpfsX32, which truncates the last 45 amino acids of the carboxyl terminal LG5 subdomain. Gene expression studies revealed that the mutant transcripts were stable and even increased, precursor laminin-332 molecules were retained intracellularly and the amount of mature extracellular heterotrimers was reduced to about 50%. Finally, the patient's keratinocytes migrated faster than normal keratinocytes. CONCLUSIONS Structural disruption of LG5 highlights the critical functions of the LG45 proteolytic region in precursor laminin-332 secretion and keratinocyte adhesion and migration. Perturbation of LG45 function might explain the non-aggressive behaviour of carcinomas in this patient.
Collapse
Affiliation(s)
- G Di Zenzo
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell'Immacolata-IRCCS, via dei Monti di Creta 104, 00167, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
A role of placental growth factor in hair growth. J Dermatol Sci 2014; 74:125-34. [DOI: 10.1016/j.jdermsci.2014.01.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 01/24/2014] [Accepted: 01/24/2014] [Indexed: 01/30/2023]
|
46
|
Lu ZX, Mao LL, Lian F, He J, Zhang WT, Dai CY, Xue S, Lu WG, Zhu HS. Cardioprotective activity of placental growth factor in a rat model of acute myocardial infarction: nanoparticle-based delivery versus direct myocardial injection. BMC Cardiovasc Disord 2014; 14:53. [PMID: 24742302 PMCID: PMC4014437 DOI: 10.1186/1471-2261-14-53] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 04/04/2014] [Indexed: 12/13/2022] Open
Abstract
Background To comparatively evaluate the cardioprotective activity of placental growth factor (PGF) delivered through direct injection and a nanoparticle-based system respectively and to study the underlying mechanisms in a rat model of acute myocardial infarction (AMI). Methods Poly lactic-co-glycolic acid (PLGA)-based PGF-carrying nanoparticles (PGF-PLGANPs) were created. The mean size and morphology of particles were analyzed with particle size analyzer and transmission electronic microscopy (TEM). Encapsulation efficiency and sustained-release dose curve were analyzed by ELISA. Sprague-Dawley rats were randomized into four groups (n = 10). While animals in the first group were left untreated as controls, those in the other 3 groups underwent surgical induction of AMI, followed by treatment with physiological saline, PGF, and PGF-PLGANPs, respectively. Cardiac function was evaluated by transthoracic echocardiography at 4 weeks after treatment. At 6 weeks, rats were sacrificed, infarction size was analyzed with Masson trichrome staining, and protein contents of TIMP-2, MT1-MMP and MMP-2 at the infarction border were determined by immunohistochemistry and western blotting analysis. Results PGF was released for at least 15 days, showing successful preparation of PGF-PLGANPs. Coronary artery ligation successfully induced AMI. Compared to physiological saline control, PGF, injected to the myocardium either as a nude molecule or in a form of nanoparticles, significantly reduced infarction size, improved cardiac function, and elevated myocardial expression of TIMP-2, MT1-MMP, and MMP-2 (P < 0.05). The effect of PGF-PLGANPs was more pronounced than that of non-encapsulated PGF (P < 0.05). Conclusion Target PGF delivery to myocardium may improve cardiac function after AMI in rats. PLGA-based nanoparticles appear to be a better approach to delivery PGF. PGF exerts its cardioprotective effect at least partially through regulating metalloproteinase-mediated myocardial tissue remodeling.
Collapse
Affiliation(s)
| | | | - Feng Lian
- Department of Cardiovascular Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu-Jian Rd, Shanghai 200127, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
47
|
Xia CY, Yu AX, Qi B, Zhou M, Li ZH, Wang WY. Analysis of blood flow and local expression of angiogenesis‑associated growth factors in infected wounds treated with negative pressure wound therapy. Mol Med Rep 2014; 9:1749-54. [PMID: 24584462 DOI: 10.3892/mmr.2014.1997] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 02/19/2014] [Indexed: 11/06/2022] Open
Abstract
Angiogenesis is involved in the wound healing process. Increased angiogenesis and blood flow constitute a major mechanism of negative pressure wound therapy (NPWT), which has been shown to facilitate the healing of infected wounds. However, the effect on the expression of angiogensis‑related growth factor remains unknown. The goal of the current study was to investigate the angiogenic factor levels prior to and following NPWT in infected wounds. A total of 20 patients with infected wounds treated with NPWT were included in the study. Patients acted as their own control; the postoperative measurements of patients were considered as the experimental group, while preoperative measurements were considered as the controlled group. Blood flow was recorded prior to and during NPWT. A total of 10 angiogensis‑related growth factors were detected using a protein biochip array to analyze the change in protein levels prior to NPWT, and on the third day during NPWT. All wounds were successfully reconstructed by skin grafting or using local flaps following NPWT. NPWT resulted in significantly increased blood flow in the wound. There was a significant increase in vascular endothelial growth factor (VEGF), EGF, platelet‑derived growth factor and angiotesin‑2 following NPWT, while basic fibroblast growth factor decreased significantly. NPWT affects the local expression of angiogenesis‑associated growth factors, which represents another mechanism to explain how NPWT accelerates wound healing.
Collapse
Affiliation(s)
- Cheng-Yan Xia
- Department of Micro‑Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ai-Xi Yu
- Department of Micro‑Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Baiwen Qi
- Department of Micro‑Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Min Zhou
- Department of Micro‑Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zong-Huan Li
- Department of Micro‑Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wei-Yang Wang
- Department of Micro‑Orthopaedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
48
|
Kent DL. Age-related macular degeneration: beyond anti-angiogenesis. Mol Vis 2014; 20:46-55. [PMID: 24426775 PMCID: PMC3888498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 01/02/2014] [Indexed: 11/03/2022] Open
Abstract
Recently, anti-vascular endothelial growth factor therapies for neovascular age-related macular degeneration have been developed. These agents, originally developed for their anti-angiogenic mechanism of action, probably also work through an anti-permeability effect in preventing or reducing the amount of leakage from submacular neovascular tissue. Other treatment modalities include laser photocoagulation, photodynamic therapy with verteporfin, and submacular surgery. In reality, these latter treatments can be similarly categorized as anti-angiogenic because their sole aim is destroying or removing choroidal neovascularization (CNV). At the cellular level, CNV resembles stereotypical tissue repair that consists of several matricellular components in addition to neovascularization. In the retina, the clinical term CNV is a misnomer since the term may more appropriately be referred to as aberrant submacular repair. Furthermore, CNV raises a therapeutic conundrum: To complete or correct any reparative process in the body, angiogenesis becomes an essential component. Anti-angiogenic therapy, in all its guises, arrests repair and causes the hypoxic environment to persist, thus fueling pro-angiogenesis and further development of CNV as a component of aberrant repair. However, we realize that anti-vascular endothelial growth factor therapy preserves vision in patients with age-related macular degeneration, albeit temporarily and therefore, repeated treatment is needed. More importantly, however, anti-angiogenic therapy demonstrates that we can at the very least tolerate neovascular tissue beneath the macula and preserve vision in contrast to our historical approach of total vascular destruction. In this clinical scenario, it may be possible to look beyond anti-angiogenesis if our goal is facilitating submacular repair without destroying the neurosensory retina. Thus, in this situation of neovascular tolerance, it may be timely to consider treatments that facilitate vascular maturation, rather than its arrest or destruction. This would neutralize hypoxia, thus removing the stimulus that drives neovascularization and in turn the need for repeated lifelong intravitreal therapy. A pro-angiogenic approach would eliminate neovascular leakage and ultimately complete repair and preserve the neurosensory retina.
Collapse
Affiliation(s)
- David L. Kent
- The Vision Clinic, Circular Road, Kilkenny, Ireland,Institute of Ageing and Chronic Disease, Faculty of Health & Life Sciences, University of Liverpool, UK
| |
Collapse
|
49
|
Van Bergen T, Jonckx B, Hollanders K, Sijnave D, Van de Velde S, Vandewalle E, Moons L, Stassen JM, Stalmans I. Inhibition of placental growth factor improves surgical outcome of glaucoma surgery. J Cell Mol Med 2013; 17:1632-43. [PMID: 24118824 PMCID: PMC3914639 DOI: 10.1111/jcmm.12151] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 09/04/2013] [Indexed: 12/28/2022] Open
Abstract
Excessive post-operative wound healing with subsequent scarring frequently leads to surgical failure of glaucoma filtration surgery (trabeculectomy). We investigated the hypothesis that placental growth factor (PlGF) plays a role in post-operative scar formation, and that it therefore may be a target for improvement of filtration surgery outcome. ELISA experiments showed that PlGF levels were significantly increased in aqueous humour of glaucoma patients and after VEGF treatment, which may indicate an important contribution of this growth factor to wound healing after trabeculectomy. Using a mouse model of glaucoma filtration surgery, we were able to show that intracameral injection of a previously characterized anti-PlGF antibody (ThromboGenics NV) significantly improved surgical outcome by increasing bleb survival and bleb area. This was associated with a significant reduction in post-operative proliferation, inflammation and angiogenesis during the first post-operative days after surgery, and with a decrease in collagen deposition at later stages. Furthermore, inhibition of PlGF seemed to be more effective than anti-VEGF-R2 treatment in improving surgical outcome, possibly via its additional effect on inflammation. These results render PlGF an appealing target for ocular wound healing and point to potential therapeutic benefits of PlGF inhibition for the prevention of surgical failure.
Collapse
|
50
|
Chua S, Sheu JJ, Chen YL, Chang LT, Sun CK, Leu S, Sung HC, Tsai TH, Chung SY, Yeh KH, Cho CL, Kao YH, Yip HK. Sitagliptin therapy enhances the number of circulating angiogenic cells and angiogenesis—evaluations in vitro and in the rat critical limb ischemia model. Cytotherapy 2013; 15:1148-63. [DOI: 10.1016/j.jcyt.2013.05.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 04/10/2013] [Accepted: 05/08/2013] [Indexed: 01/07/2023]
|