1
|
Wlodarczyk J, Leng A, Abadchi SN, Shababi N, Mokhtari-Esbuie F, Gheshlaghi S, Ravari MR, Pippenger EK, Afrasiabi A, Ha J, Abraham JM, Harmon JW. Transfection of hypoxia-inducible factor-1α mRNA upregulates the expression of genes encoding angiogenic growth factors. Sci Rep 2024; 14:6738. [PMID: 38509125 PMCID: PMC10954730 DOI: 10.1038/s41598-024-54941-w] [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: 11/08/2023] [Accepted: 02/19/2024] [Indexed: 03/22/2024] Open
Abstract
Hypoxia-Inducible Factor-1α (HIF-1α) has presented a new direction for ischemic preconditioning of surgical flaps to promote their survival. In a previous study, we demonstrated the effectiveness of HIF-1a DNA plasmids in this application. In this study, to avoid complications associated with plasmid use, we sought to express HIF-1α through mRNA transfection and determine its biological activity by measuring the upregulation of downstream angiogenic genes. We transfected six different HIF-1a mRNAs-one predominant, three variant, and two novel mutant isoforms-into primary human dermal fibroblasts using Lipofectamine, and assessed mRNA levels using RT-qPCR. At all time points examined after transfection (3, 6, and 10 h), the levels of HIF-1α transcript were significantly higher in all HIF-1α transfected cells relative to the control (all p < 0.05, unpaired Student's T-test). Importantly, the expression of HIF-1α transcription response genes (VEGF, ANG-1, PGF, FLT1, and EDN1) was significantly higher in the cells transfected with all isoforms than with the control at six and/or ten hours post-transfection. All isoforms were transfected successfully into human fibroblast cells, resulting in the rapid upregulation of all five downstream angiogenic targets tested. These findings support the potential use of HIF-1α mRNA for protecting ischemic dermal flaps.
Collapse
Affiliation(s)
- Jakub Wlodarczyk
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
- Department of General and Oncological Surgery, Medical University of Lodz, Lodz, Poland
| | - Albert Leng
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - Sanaz Nourmohammadi Abadchi
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - Niloufar Shababi
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - Farzad Mokhtari-Esbuie
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - Shayan Gheshlaghi
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - Mohsen Rouhani Ravari
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
- Department of Surgery, University of Chicago Medicine, Chicago, IL, 60637, USA
| | - Emma K Pippenger
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - Ali Afrasiabi
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - Jinny Ha
- Division of Thoracic Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - John M Abraham
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA
| | - John W Harmon
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, 1550 Orleans Street, Baltimore, MD, 21224, USA.
| |
Collapse
|
2
|
Therapeutic delivery of nucleic acids for skin wound healing. Ther Deliv 2022; 13:339-358. [PMID: 35975470 DOI: 10.4155/tde-2022-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Though wound care has advanced, treating chronic wounds remains a challenge and there are many clinical issues that must be addressed. Gene therapy is a recent approach to treating chronic wounds that remains in its developmental stage. The limited reports available describe the therapeutic applications of various forms of nucleic acid delivery for treating chronic wounds, including DNA, mRNA, siRNA, miRNA and so on. Though these bioactive molecules represent great therapeutic potential, sustaining their bioactivity in the wound bed is a challenge. To overcome this hurdle, delivery systems are also being widely investigated. In this review, nucleic acid-based therapy and its delivery for treating chronic wounds is discussed in detail.
Collapse
|
3
|
Jaurila H, Koskela M, Koivukangas V, Gäddnäs F, Salo T, Ala-Kokko TI. Growth factor expression is enhanced and extracellular matrix proteins are depressed in healing skin wounds in septic patients compared with healthy controls. APMIS 2021; 130:155-168. [PMID: 34939229 PMCID: PMC9305760 DOI: 10.1111/apm.13175] [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] [Accepted: 12/22/2021] [Indexed: 11/28/2022]
Abstract
Sepsis manifests as a dysregulated immune response to infection, damaging organs. Skin has a critical role in protecting the body. In sepsis, skin wound healing is impaired. The mechanisms behind it have been poorly studied. In this study, suction blister wounds were induced on intact abdominal skin in 15 septic patients. A single blister wound was biopsied from each patient and from 10 healthy controls. Immunohistochemical staining of growth factors and extracellular matrix (ECM) proteins was performed. Significance (p < 0.05) of the differences was calculated. The following growth factors were overexpressed in the skin of septic patients compared with healthy controls: epithelial growth factor (intact epithelium p = 0.007, migrating epithelium p = 0.038), vascular epithelial growth factor (intact epithelium p < 0.001, migrating epithelium p = 0.011) and transforming growth factor beta (migrating epithelium p = 0.002). The expression of syndecan‐1 was upregulated in the skin of septic patients compared with healthy controls (intact epithelium p = 0.048, migrating epithelium p = 0.028). The following ECM proteins had lower expression in the epithelium in septic patients than in healthy controls: tenascin‐C (migrating epithelium p = 0.03) and laminin‐332 (intact epithelium p = 0.036). In sepsis, growth factor and syndecan expression was enhanced, while ECM and basement membrane proteins were mostly depressed.
Collapse
Affiliation(s)
- Henna Jaurila
- ¹Research Group of Surgery, Anesthesia and Intensive Care, Oulu University Hospital, Oulu, Finland, Medical Research Center Oulu, University of Oulu, Finland.,Cancer and Translational Medicine Research Unit, Faculty of Medicine, Medical Research Center Oulu, University of Oulu, Finland
| | - Marjo Koskela
- ¹Research Group of Surgery, Anesthesia and Intensive Care, Oulu University Hospital, Oulu, Finland, Medical Research Center Oulu, University of Oulu, Finland
| | - Vesa Koivukangas
- ¹Research Group of Surgery, Anesthesia and Intensive Care, Oulu University Hospital, Oulu, Finland, Medical Research Center Oulu, University of Oulu, Finland
| | - Fiia Gäddnäs
- ¹Research Group of Surgery, Anesthesia and Intensive Care, Oulu University Hospital, Oulu, Finland, Medical Research Center Oulu, University of Oulu, Finland
| | - Tuula Salo
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, Medical Research Center Oulu, University of Oulu, Finland.,Research Group of Oral Health Sciences, Oulu University Hospital, Medical Research Center Oulu, University of Oulu, Finland
| | - Tero I Ala-Kokko
- ¹Research Group of Surgery, Anesthesia and Intensive Care, Oulu University Hospital, Oulu, Finland, Medical Research Center Oulu, University of Oulu, Finland
| |
Collapse
|
4
|
Chang KH, Shoureshi P, Lay F, Sebastian R, Alikhassy Habibabady Z, Born LJ, Marti GP, Meltzer SJ, Abraham JM, Harmon JW. Preconditioning of surgical pedicle flaps with DNA plasmid expressing hypoxia-inducible factor-1α (HIF-1α) promotes tissue viability. Gene Ther 2021; 28:319-328. [PMID: 33024315 DOI: 10.1038/s41434-020-00199-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 09/05/2020] [Accepted: 09/24/2020] [Indexed: 01/29/2023]
Abstract
Ischemic necrosis of surgical flaps after reconstruction is a major clinical problem. Hypoxia-inducible factor-1α (HIF-1α) is considered the master regulator of the adaptive response to hypoxia. Among its many properties, it regulates the expression of genes encoding angiogenic growth factors, which have a short half-life in vivo. To achieve a continuous application of the therapeutic, we utilized DNA plasmid delivery. Transcription of the DNA plasmid confirmed by qRT-PCR showed significantly increased mRNA for HIF-1α in the transfected tissue compared to saline control tissue. Rats were preconditioned by injecting with either HIF-1α DNA plasmid or saline intradermally in the designated flap region on each flank. Seven days after preconditioning, each rat had two isolated pedicle flaps raised with a sterile silicone sheet implanted between the skin flap and muscle layer. The flaps preconditioned with HIF-1α DNA plasmid had significantly less necrotic area. Angiogenesis measured by CD31 staining showed a significant increase in the number of vessels per high powered field in the HIF-1α group (p < 0.05). Our findings offer a potential therapeutic strategy for significantly promoting the viability of surgical pedicle flaps by ischemic preconditioning with HIF-1α DNA plasmid.
Collapse
Affiliation(s)
- Kai-Hua Chang
- Hendrix burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Pouria Shoureshi
- Hendrix burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Frank Lay
- Hendrix burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Raul Sebastian
- Department of Surgery, George Washington University School of Medicine and Health Science, Washington, DC, USA
| | | | - Louis J Born
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Guy P Marti
- Hendrix burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Clinique Saint Jean l'Ermitage, Melun, France
| | - Stephen J Meltzer
- Division of Gastroenterology, Department of Medicine and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John M Abraham
- Hendrix burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Division of Gastroenterology, Department of Medicine and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John W Harmon
- Hendrix burn and Wound Healing Laboratory, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
5
|
Gouarderes S, Doumard L, Vicendo P, Mingotaud AF, Rols MP, Gibot L. Electroporation does not affect human dermal fibroblast proliferation and migration properties directly but indirectly via the secretome. Bioelectrochemistry 2020; 134:107531. [PMID: 32335353 DOI: 10.1016/j.bioelechem.2020.107531] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 12/29/2022]
Abstract
Aesthetic wound healing is often experienced by patients after electrochemotherapy. We hypothesized that pulsed electric fields applied during electrochemotherapy (ECT) or gene electrotransfer (GET) protocols could stimulate proliferation and migration of human cutaneous cells, as described in protocols for electrostimulation of wound healing. We used videomicroscopy to monitor and quantify in real time primary human dermal fibroblast behavior when exposed in vitro to ECT and GET electric parameters, in terms of survival, proliferation and migration in a calibrated scratch wound assay. Distinct electric field intensities were applied to allow gradient in cell electropermeabilization while maintaining reversible permeabilization conditions, in order to mimic in vivo heterogeneous electric field distribution of complex tissues. Neither galvanotaxis nor statistical modification of fibroblast migration were observed in a calibrated scratch wound assay after application of ECT and GET parameters. The only effect on proliferation was observed under the strongest GET conditions, which drastically reduced the number of fibroblasts through induction of mitochondrial stress and apoptosis. Finally, we found that 24 h-conditioned cell culture medium by electrically stressed fibroblasts tended to increase the migration properties of cells that were not exposed to electric field. RT-qPCR array indicated that several growth factor transcripts were strongly modified after electroporation.
Collapse
Affiliation(s)
- Sara Gouarderes
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, France
| | - Layal Doumard
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, France; Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Patricia Vicendo
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, France
| | - Anne-Françoise Mingotaud
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, France
| | - Marie-Pierre Rols
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Laure Gibot
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, France; Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
6
|
Oryan A, Alemzadeh E, Zarei M. Basic concepts, current evidence, and future potential for gene therapy in managing cutaneous wounds. Biotechnol Lett 2019; 41:889-898. [PMID: 31256273 DOI: 10.1007/s10529-019-02701-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/19/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Several studies have investigated the role of gene therapy in the healing process. The aim of this review is to explain the gene delivery systems in wound area. RESULTS Ninety-two studies were included and comprehensively overviewed. We described the importance of viral vectors such as adenoviruses, adeno-associated viruses, and retroviruses, and conventional non-viral vectors such as naked DNA injections, liposomes, gene gun, electroporation, and nanoparticles in achieving high-level expression of genes. Application of viral transfection, liposomal vectors, and electroporation were the main gene delivery systems. Genes encoding for growth factors or cytokines have been shown to result in a better wound closure in comparison to application of the synthetic growth factors. In addition, a combination of stem cell and gene therapy has been found an effective approach in regeneration of cutaneous wounds. CONCLUSIONS This article gives an overview of the methods and investigations applied on gene therapy in wound healing. However, clinical investigations need to be undertaken to gain a better understanding of gene delivery technologies and their roles in stimulating wound repair.
Collapse
Affiliation(s)
- Ahmad Oryan
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Esmat Alemzadeh
- Department of Medical Biotechnology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Zarei
- Department of Agricultural Biotechnology, Faculty of Engineering and Technology, Imam Khomeini International University, Qazvin, Iran
| |
Collapse
|
7
|
Zhang N, Chin JS, Chew SY. Localised non-viral delivery of nucleic acids for nerve regeneration in injured nervous systems. Exp Neurol 2018; 319:112820. [PMID: 30195695 DOI: 10.1016/j.expneurol.2018.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/31/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023]
Abstract
Axons damaged by traumatic injuries are often unable to spontaneously regenerate in the adult central nervous system (CNS). Although the peripheral nervous system (PNS) has some regenerative capacity, its ability to regrow remains limited across large lesion gaps due to scar tissue formation. Nucleic acid therapy holds the potential of improving regeneration by enhancing the intrinsic growth ability of neurons and overcoming the inhibitory environment that prevents neurite outgrowth. Nucleic acids modulate gene expression by over-expression of neuronal growth factor or silencing growth-inhibitory molecules. Although in vitro outcomes appear promising, the lack of efficient non-viral nucleic acid delivery methods to the nervous system has limited the application of nucleic acid therapeutics to patients. Here, we review the recent development of efficient non-viral nucleic acid delivery platforms, as applied to the nervous system, including the transfection vectors and carriers used, as well as matrices and scaffolds that are currently used. Additionally, we will discuss possible improvements for localised nucleic acid delivery.
Collapse
Affiliation(s)
- Na Zhang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore
| | - Jiah Shin Chin
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore; NTU Institute of Health Technologies, Interdisciplinary Graduate School, Nanyang Technological University, 639798, Singapore
| | - Sing Yian Chew
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 637459, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore.
| |
Collapse
|
8
|
Sessions JW, Armstrong DG, Hope S, Jensen BD. A review of genetic engineering biotechnologies for enhanced chronic wound healing. Exp Dermatol 2018; 26:179-185. [PMID: 27574909 DOI: 10.1111/exd.13185] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2016] [Indexed: 12/29/2022]
Abstract
Traditional methods for addressing chronic wounds focus on correcting dysfunction by controlling extracellular elements. This review highlights technologies that take a different approach - enhancing chronic wound healing by genetic modification to wound beds. Featured cutaneous transduction/transfection methods include viral modalities (ie adenoviruses, adeno-associated viruses, retroviruses and lentiviruses) and conventional non-viral modalities (ie naked DNA injections, microseeding, liposomal reagents, particle bombardment and electroporation). Also explored are emerging technologies, focusing on the exciting capabilities of wound diagnostics such as pyrosequencing as well as site-specific nuclease editing tools such as CRISPR-Cas9 used to both transiently and permanently genetically modify resident wound bed cells. Additionally, new non-viral transfection methods (ie conjugated nanoparticles, multi-electrode arrays, and microfabricated needles and nanowires) are discussed that can potentially facilitate more efficient and safe transgene delivery to skin but also represent significant advances broadly to tissue regeneration research.
Collapse
Affiliation(s)
- John W Sessions
- Department of Mechanical Engineering, Brigham Young University, Provo, UT, USA
| | - David G Armstrong
- Southern Arizona Limb Salvage Alliance (SALSA), University of Arizona, Tucson, AZ, USA
| | - Sandra Hope
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Brian D Jensen
- Department of Mechanical Engineering, Brigham Young University, Provo, UT, USA
| |
Collapse
|
9
|
Jaurila H, Koivukangas V, Koskela M, Gäddnäs F, Salo S, Korvala J, Risteli M, Karhu T, Herzig KH, Salo T, Ala-Kokko TI. Inhibitory effects of serum from sepsis patients on epithelial cell migration in vitro: a case control study. J Transl Med 2017; 15:11. [PMID: 28086962 PMCID: PMC5237124 DOI: 10.1186/s12967-016-1110-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/14/2016] [Indexed: 12/05/2022] Open
Abstract
Background Sepsis delays wound re-epithelialization. In this study we explored the effect of human sepsis sera as well as the effects of cytokines, growth factors and exosomes of sepsis sera treated normal fibroblasts (NF) on keratinocyte migration and proliferation in vitro. Methods Serum samples were taken on days 1, 4, and 9 from 44 patients diagnosed with severe sepsis, and from 14 matching healthy controls. We evaluated the effects of sepsis serum with or without TNF-α, EGF, EGF receptor inhibitor or exosomes of sepsis sera treated NF on human keratinocyte (HaCaT) proliferation (BrdU assay), viability (MTT assay), and migration (horizontal wound healing model). Cytokine levels of sepsis and healthy sera were measured by multiplex assay. Comparisons between groups were carried out using SPSS statistics and P < 0.05 was considered significant. Results Severe-sepsis sera collected on days 1, 4, and 9 reduced keratinocyte proliferation by 6% (P = 0.005), 20% (P = 0.001), and 18% (P = 0.002), respectively, compared to control sera. Cell viability in cultures exposed to sepsis sera from days 4 and 9 was reduced by 38% (P = 0.01) and 58% (P < 0.001), respectively. Open-surface wounds exposed to sepsis sera from days 1 and 4 were larger than those exposed to sera from healthy controls (60 vs. 31%, P = 0.034 and 66 vs. 31%, P = 0.023, respectively). Exosomes of sepsis or healthy sera treated NF inhibited keratinocyte migration. We detected higher serum levels of cytokines TNF-α (5.7 vs. 0.7 pg/ml, P < 0.001), IL-6 (24.8 vs. 3.8 pg/ml, P < 0.001), IL-10 (30.0 vs. 11.9 pg/ml, P = 0.040), and VEGF (177.9 vs. 48.1 pg/ml, P = 0.018) in sepsis sera. Levels of EGF were significantly lower in sepsis sera than in that of healthy controls (6.5 vs. 115.6 pg/ml, P < 0.001). Sepsis serum supplemented with EGF 5 ng/ml and TNF-α in all concentrations improved keratinocyte migration. Conclusions Keratinocyte viability, proliferation and migration were reduced in severe sepsis in vitro. Exosomes from NF added in healthy or sepsis serum media inhibited keratinocyte migration. Decreased levels of EGF in sepsis sera may partially explain the delay of wound healing with severe-sepsis patients. Increased levels of TNF-α in sepsis sera do not explain diminished keratinocyte migration.
Collapse
Affiliation(s)
- Henna Jaurila
- Research Group of Surgery, Anesthesia and Intensive Care, Oulu University Hospital, P. O. Box 21, 90029, Oulu, Finland. .,Cancer and Translational Medicine Research Unit, Faculty of Medicine, Medical Research Center Oulu, University of Oulu, P.O. Box 5281, 90014, Oulu, Finland.
| | - Vesa Koivukangas
- Research Group of Surgery, Anesthesia and Intensive Care, Oulu University Hospital, P. O. Box 21, 90029, Oulu, Finland
| | - Marjo Koskela
- Research Group of Surgery, Anesthesia and Intensive Care, Oulu University Hospital, P. O. Box 21, 90029, Oulu, Finland
| | - Fiia Gäddnäs
- Research Group of Surgery, Anesthesia and Intensive Care, Oulu University Hospital, P. O. Box 21, 90029, Oulu, Finland
| | - Sirpa Salo
- Research Group of Biomedicine, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P. O. Box 5000, Oulu, 90014, Finland
| | - Johanna Korvala
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, Medical Research Center Oulu, University of Oulu, P.O. Box 5281, 90014, Oulu, Finland
| | - Maija Risteli
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, Medical Research Center Oulu, University of Oulu, P.O. Box 5281, 90014, Oulu, Finland
| | - Toni Karhu
- Research Unit of Biomedicine, Faculty of Medicine and Biocenter of Oulu, University of Oulu, P.O. Box 5000, Oulu, 90014, Finland
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine, Faculty of Medicine and Biocenter of Oulu, University of Oulu, P.O. Box 5000, Oulu, 90014, Finland.,Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, Collegium Maius, Fredry 10, 61-701, Poznan, Poland
| | - Tuula Salo
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, Medical Research Center Oulu, University of Oulu, P.O. Box 5281, 90014, Oulu, Finland.,Research Group of Oral Health Sciences, Oulu University Hospital, Medical Research Center Oulu, University of Oulu, P. O. Box 5000, Oulu, 90014, Finland
| | - Tero I Ala-Kokko
- Research Group of Surgery, Anesthesia and Intensive Care, Oulu University Hospital, P. O. Box 21, 90029, Oulu, Finland
| |
Collapse
|
10
|
Sessions JW, Hanks BW, Lindstrom DL, Hope S, Jensen BD. Transient Low-Temperature Effects on Propidium Iodide Uptake in Lance Array Nanoinjected HeLa Cells. J Nanotechnol Eng Med 2016. [DOI: 10.1115/1.4033323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Understanding environmental factors relative to transfection protocols is key for improving genetic engineering outcomes. In the following work, the effects of temperature on a nonviral transfection procedure previously described as lance array nanoinjection are examined in context of molecular delivery of propidium iodide (PI), a cell membrane impermeable nucleic acid dye, to HeLa 229 cells. For treatment samples, variables include varying the temperature of the injection solution (3C and 23C) and the magnitude of the pulsed voltage used during lance insertion into the cells (+5 V and +7 V). Results indicate that PI is delivered at levels significantly higher for samples injected at 3C as opposed to 23C at four different postinjection intervals (t = 0, 3, 6, 9 mins; p-value ≤ 0.005), reaching a maximum value of 8.3 times the positive control for 3 C/7 V pulsed samples. Suggested in this work is that between 3 and 6 mins postinjection, a large number of induced pores from the injection event close. While residual levels of PI still continue to enter the treatment samples after 6 mins, it occurs at decreased levels, suggesting from a physiological perspective that many lance array nanoinjection (LAN) induced pores have closed, some are still present.
Collapse
Affiliation(s)
- John W. Sessions
- Department of Mechanical Engineering, Brigham Young University, Provo, UT 84602 e-mail:
| | - Brad W. Hanks
- Department of Mechanical Engineering, Brigham Young University, Provo, UT 84602 e-mail:
| | - Dallin L. Lindstrom
- Department of Exercise Science, Brigham Young University, Provo, UT 84602 e-mail:
| | - Sandra Hope
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602 e-mail:
| | - Brian D. Jensen
- Department of Mechanical Engineering, Brigham Young University, Provo, UT 84602 e-mail:
| |
Collapse
|
11
|
Gibot L, Rols MP. Gene transfer by pulsed electric field is highly promising in cutaneous wound healing. Expert Opin Biol Ther 2015; 16:67-77. [DOI: 10.1517/14712598.2016.1098615] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
12
|
Fang YL, Chen XG, W T G. Gene delivery in tissue engineering and regenerative medicine. J Biomed Mater Res B Appl Biomater 2014; 103:1679-99. [PMID: 25557560 DOI: 10.1002/jbm.b.33354] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 11/07/2014] [Accepted: 11/18/2014] [Indexed: 12/13/2022]
Abstract
As a promising strategy to aid or replace tissue/organ transplantation, gene delivery has been used for regenerative medicine applications to create or restore normal function at the cell and tissue levels. Gene delivery has been successfully performed ex vivo and in vivo in these applications. Excellent proliferation capabilities and differentiation potentials render certain cells as excellent candidates for ex vivo gene delivery for regenerative medicine applications, which is why multipotent and pluripotent cells have been intensely studied in this vein. In this review, gene delivery is discussed in detail, along with its applications to tissue engineering and regenerative medicine. A definition of a stem cell is compared to a definition of a stem property, and both provide the foundation for an in-depth look at gene delivery investigations from a germ lineage angle.
Collapse
Affiliation(s)
- Y L Fang
- Department of Chemical & Biomolecular Engineering, Laboratory for Gene Therapy and Cellular Engineering, Tulane University, 300 Lindy Boggs Center, New Orleans, Louisiana, 70118
| | - X G Chen
- Department of Chemical & Biomolecular Engineering, Laboratory for Gene Therapy and Cellular Engineering, Tulane University, 300 Lindy Boggs Center, New Orleans, Louisiana, 70118
| | - Godbey W T
- Department of Chemical & Biomolecular Engineering, Laboratory for Gene Therapy and Cellular Engineering, Tulane University, 300 Lindy Boggs Center, New Orleans, Louisiana, 70118
| |
Collapse
|
13
|
Finch PW, Mark Cross LJ, McAuley DF, Farrell CL. Palifermin for the protection and regeneration of epithelial tissues following injury: new findings in basic research and pre-clinical models. J Cell Mol Med 2014; 17:1065-87. [PMID: 24151975 PMCID: PMC4118166 DOI: 10.1111/jcmm.12091] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 05/06/2013] [Accepted: 05/15/2013] [Indexed: 02/06/2023] Open
Abstract
Keratinocyte growth factor (KGF) is a paracrine-acting epithelial mitogen produced by cells of mesenchymal origin, that plays an important role in protecting and repairing epithelial tissues. Pre-clinical data initially demonstrated that a recombinant truncated KGF (palifermin) could reduce gastrointestinal injury and mortality resulting from a variety of toxic exposures. Furthermore, the use of palifermin in patients with hematological malignancies reduced the incidence and duration of severe oral mucositis experienced after intensive chemoradiotherapy. Based upon these findings, as well as the observation that KGF receptors are expressed in many, if not all, epithelial tissues, pre-clinical studies have been conducted to determine the efficacy of palifermin in protecting different epithelial tissues from toxic injury in an attempt to model various clinical situations in which it might prove to be of benefit in limiting tissue damage. In this article, we review these studies to provide the pre-clinical background for clinical trials that are described in the accompanying article and the rationale for additional clinical applications of palifermin.
Collapse
|
14
|
Dou C, Lay F, Ansari AM, Rees DJ, Ahmed AK, Kovbasnjuk O, Matsangos AE, Du J, Hosseini SM, Steenbergen C, Fox-Talbot K, Tabor AT, Williams JA, Liu L, Marti GP, Harmon JW. Strengthening the skin with topical delivery of keratinocyte growth factor-1 using a novel DNA plasmid. Mol Ther 2014; 22:752-61. [PMID: 24434934 PMCID: PMC3982499 DOI: 10.1038/mt.2014.2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 12/23/2013] [Indexed: 12/25/2022] Open
Abstract
Fragile skin, susceptible to decubitus ulcers and incidental trauma, is a problem particularly for the elderly and for those with spinal cord injury. Here, we present a simple approach to strengthen the skin by the topical delivery of keratinocyte growth factor-1 (KGF-1) DNA. In initial feasibility studies with the novel minimalized, antibiotic-free DNA expression vector, NTC8385-VA1, the reporter genes luciferase and enhanced green fluorescent protein were delivered. Transfection was documented when luciferase expression significantly increased after transfection. Microscopic imaging of enhanced green fluorescent protein-transfected skin showed green fluorescence in hair follicles, hair shafts, and dermal and superficial epithelial cells. With KGF-1 transfection, KGF-1 mRNA level and protein production were documented with quantitative reverse transcriptase-polymerase chain reaction and immunohistochemistry, respectively. Epithelial thickness of the transfected skin in the KGF group was significantly increased compared with the control vector group (26 ± 2 versus 16 ± 4 µm) at 48 hours (P = 0.045). Dermal thickness tended to be increased in the KGF group (255 ± 36 versus 162 ± 16 µm) at 120 hours (P = 0.057). Biomechanical assessment showed that the KGF-1-treated skin was significantly stronger than control vector-transfected skin. These findings indicate that topically delivered KGF-1 DNA plasmid can increase epithelial thickness and strength, demonstrating the potential of this approach to restore compromised skin.
Collapse
Affiliation(s)
- Chunqing Dou
- 1] Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China [2] Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Frank Lay
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amir Mehdi Ansari
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Donald J Rees
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ali Karim Ahmed
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Olga Kovbasnjuk
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aerielle E Matsangos
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Junkai Du
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sayed Mohammad Hosseini
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Charles Steenbergen
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Karen Fox-Talbot
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | - Lixin Liu
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guy P Marti
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John W Harmon
- Department of Surgery and Hendrix Burn/Wound Laboratory, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
15
|
|
16
|
An electrospun scaffold integrating nucleic acid delivery for treatment of full-thickness wounds. Biomaterials 2013; 34:3891-901. [PMID: 23453058 DOI: 10.1016/j.biomaterials.2013.02.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 02/06/2013] [Indexed: 11/23/2022]
Abstract
We developed a multi-functional construct capable of controlled delivery of bioactive substances that can improve wound repair by supporting the intrinsic ability of the skin to heal. We synthesized electrospun scaffolds-composed of a blend of the degradable polymers poly(l-lactide) (PLA) or polycaprolactone (PCL)-that produce highly efficient non-viral in vivo gene delivery to cells in the wound bed, provide a protective barrier during early wound healing, and support cell migration and growth. This multi-functional material was tested for its influence on wound healing: scaffolds were loaded with plasmids encoding keratinocyte growth factor (KGF) and applied to full-thickness wounds in mice. Compared to scaffolds with control plasmids, animals receiving the KGF plasmid-loaded scaffold produced significant enhancements in wound healing, which was quantified by improvements in the rate of wound re-epithelialization, keratinocyte proliferation, and granulation response. Further, we quantified the expression level of endogenous and plasmid-derived KGF in wound samples: qRT-PCR on wound sections revealed a correlation between the levels of plasmid-derived protein expression and histological analysis of wound healing, revealing an inverse relationship between the expression level of exogenous KGF and the size of the unhealed epithelial layer in wounds. Our findings suggest that engineered nanofiber PLA/PCL scaffolds are capable of highly efficient controlled DNA delivery and are promising materials for treatment of cutaneous wounds.
Collapse
|
17
|
Acute and impaired wound healing: pathophysiology and current methods for drug delivery, part 2: role of growth factors in normal and pathological wound healing: therapeutic potential and methods of delivery. Adv Skin Wound Care 2012; 25:349-70. [PMID: 22820962 DOI: 10.1097/01.asw.0000418541.31366.a3] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This is the second of 2 articles that discuss the biology and pathophysiology of wound healing, reviewing the role that growth factors play in this process and describing the current methods for growth factor delivery into the wound bed.
Collapse
|
18
|
Common healthcare challenges. PLASMA MEDICINE 2012. [DOI: 10.1017/cbo9780511902598.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
19
|
Schreml S, Szeimies RM, Prantl L, Landthaler M, Babilas P. Wound healing in the 21st century. J Am Acad Dermatol 2010; 63:866-81. [DOI: 10.1016/j.jaad.2009.10.048] [Citation(s) in RCA: 194] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 10/23/2009] [Accepted: 10/23/2009] [Indexed: 01/13/2023]
|
20
|
Therapeutic levels of erythropoietin (EPO) achieved after gene electrotransfer to skin in mice. Gene Ther 2010; 17:1077-84. [PMID: 20410932 DOI: 10.1038/gt.2010.46] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Gene electrotransfer refers to gene transfection by electroporation and is an effective non-viral method for delivering naked DNA into cells and tissues. This study presents data from gene electrotransfer with erythropoietin (EPO) to mouse skin. Nine-week-old female NMRI mice received one, two or three intradermal injections of 50 microg EPO plasmid and were subsequently electroporated. With plate electrodes and 100 microg of EPO, a significant increase in hemoglobin (P<0.01) was observed compared with controls. The level of hemoglobin peaked after 5 weeks but stayed significantly elevated for more than 3 months. Serum EPO was significantly increased (P<0.001) 24 h after the transfection and remained significantly different compared with controls until the maximum level of serum EPO was reached after 2 weeks. Eight weeks after the transfection serum EPO returned to baseline. In this study, we have established that gene electrotransfer to skin of even small amounts of DNA can lead to systemically therapeutic levels of protein. This means that in addition to DNA vaccinations, there is a potential utility for electroporation in alleviating systemic diseases such as cancer and protein deficiency disorders.
Collapse
|
21
|
Ferraro B, Cruz YL, Baldwin M, Coppola D, Heller R. Increased perfusion and angiogenesis in a hindlimb ischemia model with plasmid FGF-2 delivered by noninvasive electroporation. Gene Ther 2010; 17:763-9. [PMID: 20393507 DOI: 10.1038/gt.2010.43] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Gene therapy approaches delivering fibroblast growth factor-2 (FGF-2) have shown promise as a potential treatment for increasing blood flow to ischemic limbs. Currently, effective noninvasive techniques to deliver plasmids encoding genes of therapeutic interest, such as FGF-2, are limited. We sought to determine if intradermal injection of plasmid DNA encoding FGF-2 (pFGF) followed by noninvasive cutaneous electroporation (pFGFE+) could increase blood flow and angiogenesis in a rat model of hindlimb ischemia. pFGFE+ or control treatments were administered on postoperative day 0. Compared to injection of pFGF alone (pFGFE-), delivery of pFGFE+ significantly increased FGF-2 expression for 10 days. Further, the increase in FGF-2 expression with pFGFE+ was sufficient to significantly increase ischemic limb blood flow, measured by laser Doppler perfusion imaging, beginning on postoperative day 3. Ischemic limb blood flow in the pFGFE+ treatment group remained significantly higher than all control groups through the end point of the study, postoperative day 14. Immunohistochemical staining of gastrocnemius cross sections determined there was a twofold increase in capillary density in the pFGFE+ treatment group. Our results suggest that pFGFE+ is a potential noninvasive, nonviral therapeutic approach to increase perfusion and angiogenesis for the treatment of limb ischemia.
Collapse
Affiliation(s)
- B Ferraro
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA
| | | | | | | | | |
Collapse
|
22
|
Abstract
Abnormal wound healing is a major complication of both type 1 and type 2 diabetes, with nonhealing foot ulcerations leading in the worst cases to lower-limb amputation. Wound healing requires the integration of complex cellular and molecular events in successive phases of inflammation, cell proliferation, cell migration, angiogenesis and re-epithelialisation. A link between wound healing and the nervous system is clinically apparent as peripheral neuropathy is reported in 30-50% of diabetic patients and is the most common and sensitive predictor of foot ulceration. Indeed, a bidirectional connection between the nervous and the immune systems and its role in wound repair has emerged as one of the focal features of the wound-healing dogma. This review provides a broad overview of the mediators of this connection, which include neuropeptides and cytokines released from nerve fibres, immune cells and cutaneous cells. In-depth understanding of the signalling pathways in the neuroimmune axis in diabetic wound healing is vital to the development of successful wound-healing therapies.
Collapse
|
23
|
Heller LC, Jaroszeski MJ, Coppola D, Heller R. Comparison of electrically mediated and liposome-complexed plasmid DNA delivery to the skin. GENETIC VACCINES AND THERAPY 2008; 6:16. [PMID: 19055808 PMCID: PMC2631522 DOI: 10.1186/1479-0556-6-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Accepted: 12/04/2008] [Indexed: 01/27/2023]
Abstract
BACKGROUND Electroporation is an established technique for enhancing plasmid delivery to many tissues in vivo, including the skin. We have previously demonstrated efficient delivery of plasmid DNA to the skin utilizing a custom-built four-plate electrode. The experiments described here further evaluate cutaneous plasmid delivery using in vivo electroporation. Plasmid expression levels are compared to those after liposome mediated delivery. METHODS Enhanced electrically-mediated delivery, and less extensively, liposome complexed delivery, of a plasmid encoding the reporter luciferase was tested in rodent skin. Expression kinetics and tissue damage were explored as well as testing in a second rodent model. RESULTS Experiments confirm that electroporation alone is more effective in enhancing reporter gene expression than plasmid injection alone, plasmid conjugation with liposomes followed by injection, or than the combination of liposomes and electroporation. However, with two time courses of multiple electrically-mediated plasmid deliveries, neither the levels nor duration of transgene expression are significantly increased. Tissue damage may increase following a second treatment, no further damage is observed after a third treatment. When electroporation conditions utilized in a mouse model are tested in thicker rat skin, only higher field strengths or longer pulses were as effective in plasmid delivery. CONCLUSION Electroporation enhances reporter plasmid delivery to the skin to a greater extent than the liposome conjugation method tested. Multiple deliveries do not necessarily result in higher or longer term expression. In addition, some impact on tissue integrity with respect to surface damage is observed. Pulsing conditions should be optimized for the model and for the expression profile desired.
Collapse
Affiliation(s)
- Loree C Heller
- Center for Molecular Delivery, University of South Florida, Tampa, FL, USA.
| | | | | | | |
Collapse
|
24
|
Liu L, Marti GP, Wei X, Zhang X, Zhang H, Liu YV, Nastai M, Semenza GL, Harmon JW. Age-dependent impairment of HIF-1alpha expression in diabetic mice: Correction with electroporation-facilitated gene therapy increases wound healing, angiogenesis, and circulating angiogenic cells. J Cell Physiol 2008; 217:319-27. [PMID: 18506785 DOI: 10.1002/jcp.21503] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Wound healing is impaired in elderly patients with diabetes mellitus. We hypothesized that age-dependent impairment of cutaneous wound healing in db/db diabetic mice: (a) would correlate with reduced expression of the transcription factor hypoxia-inducible factor 1alpha (HIF-1alpha) as well as its downstream target genes; and (b) could be overcome by HIF-1alpha replacement therapy. Wound closure, angiogenesis, and mRNA expression in excisional skin wounds were analyzed and circulating angiogenic cells (CACs) were quantified in db/db mice that were untreated or received electroporation-facilitated HIF-1alpha gene therapy. HIF-1alpha mRNA levels in wound tissue were significantly reduced in older (4-6 months) as compared to younger (1.5-2 months) db/db mice. Expression of mRNAs encoding the angiogenic cytokines vascular endothelial growth factor (VEGF), angiopoietin 1 (ANGPT1), ANGPT2, platelet-derived growth factor B (PDGF-B), and placental growth factor (PLGF) was also impaired in wounds of older db/db mice. Intradermal injection of plasmid gWIZ-CA5, which encodes a constitutively active form of HIF-1alpha, followed by electroporation, induced increased levels of HIF-1alpha mRNA at the injection site on day 3 and increased levels of VEGF, PLGF, PDGF-B, and ANGPT2 mRNA on day 7. CACs in peripheral blood increased 10-fold in mice treated with gWIZ-CA5. Wound closure was significantly accelerated in db/db mice treated with gWIZ-CA5 as compared to mice treated with empty vector. Thus, HIF-1alpha gene therapy corrects the age-dependent impairment of HIF-1alpha expression, angiogenic cytokine expression, and CACs that contribute to the age-dependent impairment of wound healing in db/db mice.
Collapse
Affiliation(s)
- Lixin Liu
- Section of Surgical Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Wound infection has always been an unconquered problem temporarily improved with the discovery of antibiotics but now struggling with an epidemic of resistant organisms. Wound healing has become a popular sub-speciality for the doctor and allied health professional working in the laboratory or at the bedside. It is a field with many new journals and frequent congresses that publish elaborate proceedings. These have been examined for this review. RECENT FINDINGS Measurement of infection remains elusive. A clear difference between contamination and infection is difficult to delineate. In the era of the HIV/AIDS epidemic attention is drawn to host factors, which when attended to are as effective in suppressing infection as antisepsis. The bacterial capacity to put on a protective coat known as biofilm is a newly investigated system. SUMMARY Antisepsis and antibiotics continue to provide a wealth of studies and some new technology. The evidence base, as examined by Cochrane systematic reviews, suggests that our strategy for preventing and dealing with infection of wounds needs further refinement.
Collapse
Affiliation(s)
- Terence J Ryan
- Oxford International Wound Healing Foundation, Oxford, UK.
| |
Collapse
|