1
|
Sumpio BJ, Dallas A, Berger AG, Li Z, Wang E, Mezghani I, Contreras M, Theocharidis G, Ilves H, Hammond PT, Johnston BH, Veves A. Use of Therapeutic RNAs to Accelerate Wound Healing in Diabetic Rabbit Wounds. Adv Wound Care (New Rochelle) 2024; 13:435-445. [PMID: 38183631 PMCID: PMC11535488 DOI: 10.1089/wound.2023.0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024] Open
Abstract
Introduction: Diabetes mellitus (DM) affects over 422 million people globally. Patients with DM are subject to a myriad of complications, of which diabetic foot ulcers (DFUs) are the most common with ∼25% chance of developing these wounds throughout their lifetime. Innovation: Currently there are no therapeutic RNAs approved for use in DFUs. Use of dressings containing novel layer-by-layer (LbL)-formulated therapeutic RNAs that inhibit PHD2 and miR-210 can significantly improve diabetic wound healing. These dressings provide sustained release of therapeutic RNAs to the wounds locally without systemic side effects. Clinical Problem Addressed: Diabetic foot wounds are difficult to heal and often result in significant patient morbidity and mortality. Materials and Methods: We used the diabetic neuroischemic rabbit model of impaired wound healing. Diabetes was induced in the rabbits with alloxan, and neuroischemia was induced by ligating the central neurovascular bundle of each ear. Four 6-mm full-thickness wounds were created on each ear. A LbL technique was used to conformally coat the wound dressings with chemically modified RNAs, including an antisense oligonucleotide (antimiR) targeting microRNA-210 (miR-210), an short synthetic hairpin RNA (sshRNA) targeting PHD2, or both. Results: Wound healing was improved by the antimiR-210 but not the PHD2-sshRNA. Specific knockdown of miR-210 in tissue as measured by RT-qPCR was ∼8 Ct greater than nonspecific controls, and this apparent level of knockdown (>99%) suggests that delivery to the tissue is highly efficient at the administered dose. Discussion: Healing of ischemic/neuropathic wounds in diabetic rabbits was accelerated upon inhibition of miR-210 by LbL delivery to the wound bed. miR-210 inhibition was achieved using a chemically modified antisense RNA.
Collapse
Affiliation(s)
- Brandon J. Sumpio
- Rongxiang Xu Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Anne Dallas
- SomaGenics, Inc., Santa Cruz, California, USA
| | - Adam G. Berger
- Harvard-MIT Division of Health Sciences and Technology, Department of Chemical Engineering, Institute for Soldier Nanotechnologies, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Zhuqing Li
- Rongxiang Xu Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Enya Wang
- Rongxiang Xu Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ikram Mezghani
- Rongxiang Xu Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mauricio Contreras
- Rongxiang Xu Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Georgios Theocharidis
- Rongxiang Xu Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Heini Ilves
- SomaGenics, Inc., Santa Cruz, California, USA
| | - Paula T. Hammond
- Harvard-MIT Division of Health Sciences and Technology, Department of Chemical Engineering, Institute for Soldier Nanotechnologies, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Aristidis Veves
- Rongxiang Xu Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Bonnici L, Suleiman S, Schembri-Wismayer P, Cassar A. Targeting Signalling Pathways in Chronic Wound Healing. Int J Mol Sci 2023; 25:50. [PMID: 38203220 PMCID: PMC10779022 DOI: 10.3390/ijms25010050] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Chronic wounds fail to achieve complete closure and are an economic burden to healthcare systems due to the limited treatment options and constant medical attention. Chronic wounds are characterised by dysregulated signalling pathways. Research has focused on naturally derived compounds, stem-cell-based therapy, small molecule drugs, oligonucleotide delivery nanoparticles, exosomes and peptide-based platforms. The phosphoinositide-3-kinase (PI3K)/protein kinase B (AKT), Wingless-related integration (Wnt)/β-catenin, transforming growth factor-β (TGF-β), nuclear factor erythroid 2-related factor 2 (Nrf2), Notch and hypoxia-inducible factor 1 (HIF-1) signalling pathways have critical roles in wound healing by modulating the inflammatory, proliferative and remodelling phases. Moreover, several regulators of the signalling pathways were demonstrated to be potential treatment targets. In this review, the current research on targeting signalling pathways under chronic wound conditions will be discussed together with implications for future studies.
Collapse
Affiliation(s)
| | | | | | - Analisse Cassar
- Department of Anatomy, University of Malta, MSD 2080 Msida, Malta; (L.B.); (S.S.); (P.S.-W.)
| |
Collapse
|
3
|
Shaabani E, Sharifiaghdam M, Faridi-Majidi R, De Smedt SC, Braeckmans K, Fraire JC. Gene therapy to enhance angiogenesis in chronic wounds. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 29:871-899. [PMID: 36159590 PMCID: PMC9464651 DOI: 10.1016/j.omtn.2022.08.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Skin injuries and chronic non-healing wounds are one of the major global burdens on the healthcare systems worldwide due to their difficult-to-treat nature, associated co-morbidities, and high health care costs. Angiogenesis has a pivotal role in the wound-healing process, which becomes impaired in many chronic non-healing wounds, leading to several healing disorders and complications. Therefore, induction or promotion of angiogenesis can be considered a promising approach for healing of chronic wounds. Gene therapy is one of the most promising upcoming strategies for the treatment of chronic wounds. It can be classified into three main approaches: gene augmentation, gene silencing, and gene editing. Despite the increasing number of encouraging results obtained using nucleic acids (NAs) as active pharmaceutical ingredients of gene therapy, efficient delivery of NAs to their site of action (cytoplasm or nucleus) remains a key challenge. Selection of the right therapeutic cargo and delivery methods is crucial for a favorable prognosis of the healing process. This article presents an overview of gene therapy and non-viral delivery methods for angiogenesis induction in chronic wounds.
Collapse
|
4
|
Song J, Hu L, Liu B, Jiang N, Huang H, Luo J, Wang L, Zeng J, Huang F, Huang M, Cai L, Tang L, Chen S, Chen Y, Wu A, Zheng S, Chen Q. The Emerging Role of Immune Cells and Targeted Therapeutic Strategies in Diabetic Wounds Healing. J Inflamm Res 2022; 15:4119-4138. [PMID: 35898820 PMCID: PMC9309318 DOI: 10.2147/jir.s371939] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/07/2022] [Indexed: 11/23/2022] Open
Abstract
Poor wound healing in individuals with diabetes has long plagued clinicians, and immune cells play key roles in the inflammation, proliferation and remodeling that occur in wound healing. When skin integrity is damaged, immune cells migrate to the wound bed through the actions of chemokines and jointly restore tissue homeostasis and barrier function by exerting their respective biological functions. An imbalance of immune cells often leads to ineffective and disordered inflammatory responses. Due to the maladjusted microenvironment, the wound is unable to smoothly transition to the proliferation and remodeling stage, causing it to develop into a chronic refractory wound. However, chronic refractory wounds consistently lead to negative outcomes, such as long treatment cycles, high hospitalization rates, high medical costs, high disability rates, high mortality rates, and many adverse consequences. Therefore, strategies that promote the rational distribution and coordinated development of immune cells during wound healing are very important for the treatment of diabetic wounds (DW). Here, we explored the following aspects by performing a literature review: 1) the current situation of DW and an introduction to the biological functions of immune cells; 2) the role of immune cells in DW; and 3) existing (or undeveloped) therapies targeting immune cells to promote wound healing to provide new ideas for basic research, clinical treatment and nursing of DW.
Collapse
Affiliation(s)
- Jianying Song
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
| | - Lixin Hu
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
| | - Bo Liu
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
| | - Nan Jiang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
| | - Houqiang Huang
- Department of Nursing, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - JieSi Luo
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
| | - Long Wang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
| | - Jing Zeng
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
| | - Feihong Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
| | - Min Huang
- Department of Respiratory and Critical Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Luyao Cai
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
| | - Lingyu Tang
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
| | - Shunli Chen
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
| | - Yinyi Chen
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
| | - Anguo Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
| | - Silin Zheng
- Department of Nursing, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Qi Chen
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- School of Nursing, Southwest Medical University, Luzhou, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| |
Collapse
|
5
|
Zhu J, Chen C, Dong J, Cheng S, Li G, Wang C, Ouyang D, Leung CH, Lin L. Artificial intelligence-aided discovery of prolyl hydroxylase 2 inhibitors to stabilize hypoxia inducible factor-1α and promote angiogenesis. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
6
|
HIF1A promotes miR-210/miR-424 transcription to modulate the angiogenesis in HUVECs and HDMECs via sFLT1 under hypoxic stress. Mol Cell Biochem 2022; 477:2107-2119. [PMID: 35488146 DOI: 10.1007/s11010-022-04428-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 03/30/2022] [Indexed: 01/08/2023]
Abstract
Angiogenesis is a critical process during human skin wound healing. However, hypoxia might lead to the dysfunction of the cellular interplay of endothelial cells and subcutaneous fibroblasts, resulting in the deregulation of angiogenesis. HIF1A is a key regulatory of the recovery of intracellular homeostasis under hypoxia. In the present study, the detailed role and mechanism of HIF1A in the angiogenesis under hypoxia were investigated. Via bioinformatic analyses on microarray profiles (GSE1041 and GSE17944), solube fms-related tyrosine kinase 1 (sFLT1, also known as sVEGFR1) and miR-210/miR-424 might be involved in HIF1A function on the angiogenesis under hypoxia in human umbilical vascular endothelium cells (HUVECs) and human dermal microvascular endothelial cells (HDMECs). In the present study, we identified sFLT1 as a downregulated gene in response to hypoxia and HIF1A overexpression in HUVECs and HDMECs. sFLT1 overexpression inhibited the capacity of migration and angiogenesis and significantly reversed the inducible effects of HIF1A on the migration and angiogenesis in both cell lines. miR-210 and miR-424 were upregulated by hypoxia and targeted sFLT1 3'-UTR to negatively modulate its expression. HIF1A modulated sFLT1 expression, VEGF signaling, and the migration and angiogenesis in HUVECs and HDMECs via miR-210/miR-424. Regarding the molecular mechanism, HIF1A bound the promoter region of miR-210 and miR-424 to activate their transcription, while miR-210/miR-424 bound sFLT1 3'-UTR to suppress its expression. In summary, HIF1A/miR-210/miR-424/sFLT1 axis modulates the angiogenesis in HUVECs and HDMECs upon hypoxic condition via VEGF signaling.
Collapse
|
7
|
Ikeda T, Nakamura K, Kida T, Oku H. Possible roles of anti-type II collagen antibody and innate immunity in the development and progression of diabetic retinopathy. Graefes Arch Clin Exp Ophthalmol 2022; 260:387-403. [PMID: 34379187 PMCID: PMC8786754 DOI: 10.1007/s00417-021-05342-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 11/08/2022] Open
Abstract
The pathogenesis of both diabetic retinopathy (DR) and rheumatoid arthritis (RA) has recently been considered to involve autoimmunity. Serum and synovial fluid levels of anti-type II collagen antibodies increase early after the onset of RA, thus inducing immune responses and subsequent hydrarthrosis and angiogenesis, which resemble diabetic macular edema and proliferative DR (PDR), respectively. We previously reported that DR is also associated with increased serum levels of anti-type II collagen antibodies. Retinal hypoxia in DR may induce pericytes to express type II collagen, resulting in autoantibody production against type II collagen. As the result of blood-retinal barrier disruption, anti-type II collagen antibodies in the serum come into contact with type II collagen around the retinal vessels. A continued loss of pericytes and type II collagen around the retinal vessels may result in a shift of the immune reaction site from the retina to the vitreous. It has been reported that anti-inflammatory M2 macrophages increased in the vitreous of PDR patients, accompanied by the activation of the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a key regulator of innate immunity. M2 macrophages promote angiogenesis and fibrosis, which might be exacerbated and prolonged by dysregulated innate immunity.
Collapse
Affiliation(s)
- Tsunehiko Ikeda
- Department of Ophthalmology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan.
- Department of Ophthalmology, Osaka Kaisei Hospital, 1-6-10 Miyahara Yodogawa-ku, Osaka City, Osaka, Japan.
| | | | - Teruyo Kida
- Department of Ophthalmology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| | - Hidehiro Oku
- Department of Ophthalmology, Osaka Medical and Pharmaceutical University, Takatsuki City, Osaka, Japan
| |
Collapse
|
8
|
Berger AG, Chou JJ, Hammond PT. Approaches to Modulate the Chronic Wound Environment Using Localized Nucleic Acid Delivery. Adv Wound Care (New Rochelle) 2021; 10:503-528. [PMID: 32496978 PMCID: PMC8260896 DOI: 10.1089/wound.2020.1167] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
Significance: Nonhealing wounds have been the subject of decades of basic and clinical research. Despite new knowledge about the biology of impaired wound healing, little progress has been made in treating chronic wounds, leaving patients with few therapeutic options. Diabetic ulcers are a particularly common form of nonhealing wound. Recent Advances: Recently, investigation of therapeutic nucleic acids (TNAs), including plasmid DNA, small interfering RNA, microRNA mimics, anti-microRNA oligonucleotides, messenger RNA, and antisense oligonucleotides, has created a new treatment strategy for chronic wounds. TNAs can modulate the wound toward a prohealing environment by targeting gene pathways associated with inflammation, proteases, cell motility, angiogenesis, epithelialization, and oxidative stress. A variety of delivery systems have been investigated for TNAs, including dendrimers, lipid nanoparticles (NPs), polymeric micelles, polyplexes, metal NPs, and hydrogels. This review summarizes recent developments in TNA delivery for therapeutic targets associated with chronic wounds, with an emphasis on diabetic ulcers. Critical Issues: Translational potential of TNAs remains a key challenge; we highlight some drug delivery approaches for TNAs that may hold promise. We also describe current commercial efforts to locally deliver nucleic acids to modulate the wound environment. Future Directions: Localized nucleic acid delivery holds promise for the treatment of nonhealing chronic wounds. Future efforts to improve targeting of these nucleic acid therapies in the wound with both spatial and temporal control through drug delivery systems will be crucial to successful clinical translation.
Collapse
Affiliation(s)
- Adam G. Berger
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jonathan J. Chou
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Paula T. Hammond
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
9
|
Sun Y, Xiong X, Wang X. HIF1α/miR-199a/ADM feedback loop modulates the proliferation of human dermal microvascular endothelial cells (HDMECs) under hypoxic condition. Cell Cycle 2019; 18:2998-3009. [PMID: 31537150 DOI: 10.1080/15384101.2019.1666611] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Hypoxia-inducible factor 1α (HIF1α) plays a protective role in the hypoxia-induced cellular injury. In the present study, we attempted to investigate the role and mechanism of HIF1αin human dermal microvascular endothelial cells (hDMECs), a common-used cell model for researches on the hypoxia-induced injury during skin wounds healing. As revealed by ChIP and online tools prediction and confirmed by luciferase reporter and ChIP assays, HIF1A can bind to the promoter regions of ADM and miR-199a, while miR-199a directly binds to the 3'UTR of HIF1A and ADM. Hypoxia stress induces HIF1α and ADM expression while inhibits miR-199a expression. Under hypoxic condition, HIF1α knockdown increases the nucleus translocation of p65 and the release of TNF-α and IL-8, inhibits the proliferation and migration, while promotes the cellular permeability in HDMECs upon hypoxic stress, while ADM overexpression and miR-199a inhibition exerted an opposite effect on HDMECs. ADM overexpression or miR-199a inhibition could partially reverse the effect of HIF1A knockdown under hypoxia. In summary, we demonstrate a feedback loop consists of HIF1α, miR-199a, and ADM which protect HDMECs from hypoxia-induced cellular injury by modulating the inflammation response, cell proliferation, migration and permeability in HDMECs.
Collapse
Affiliation(s)
- Yang Sun
- Department of plastic surgery and burns surgery, The Second Xiangya Hospital, Central South University , Changsha , Hunan , China
| | - Xiang Xiong
- Department of plastic surgery and burns surgery, The Second Xiangya Hospital, Central South University , Changsha , Hunan , China
| | - Xiancheng Wang
- Department of plastic surgery and burns surgery, The Second Xiangya Hospital, Central South University , Changsha , Hunan , China
| |
Collapse
|
10
|
Qiu S, Jia Y, Sun Y, Han P, Xu J, Wen G, Chai Y. Von Hippel-Lindau (VHL) Protein Antagonist VH298 Improves Wound Healing in Streptozotocin-Induced Hyperglycaemic Rats by Activating Hypoxia-Inducible Factor- (HIF-) 1 Signalling. J Diabetes Res 2019; 2019:1897174. [PMID: 30911550 PMCID: PMC6398031 DOI: 10.1155/2019/1897174] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 11/01/2018] [Accepted: 11/29/2018] [Indexed: 01/04/2023] Open
Abstract
AIMS The purpose of the present research is to investigate the effects of the VHL protein antagonist, VH298, on functional activities of fibroblasts and vascular endothelial cells and the effects on the wound healing process in a streptozotocin-induced hyperglycaemic rat model. METHODS HIF-1α and hydroxy-HIF-1α protein levels in VH298-treated rat fibroblasts (rFb) were measured by immunoblotting, rFb proliferation was detected by the CCK-8 assay, and mRNA levels of related genes were measured by quantitative RT-PCR. In vitro wound healing was simulated by the scratch test; angiogenesis was measured by the human umbilical vein endothelial cell (hUVEC) tube formation assay. VH298 or PBS was locally injected into wounds in rat models with streptozotocin- (STZ-) induced hyperglycaemia, the wound tissues were harvested, and haematoxylin-eosin (HE) and Masson trichrome staining and immunohistochemical processes were conducted. RESULTS HIF-1α and hydroxy-HIF-1α levels increased in VH298-treated rFb, in a time- and dose-dependent manner. Thirty micromolar VH298 could significantly increase cell proliferation, angiogenesis, and gene expression of type I collagen-α1 (Col1-α1), vascular endothelial growth factor A (VEGF-A), and insulin-like growth factor 1 (IGF-1). The VH298-treated wound had a better healing pattern, activation of HIF-1 signalling, and vascularization. CONCLUSIONS Taken together, VH298 activated the HIF-1 signalling pathway by stabilizing both HIF-1α and hydroxy-HIF-1α. VH298 enhanced rFb functions, promoted hUVEC angiogenesis, and accelerated wound healing in the rat model mimicking diabetes mellitus.
Collapse
Affiliation(s)
- Shuo Qiu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yachao Jia
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yunchu Sun
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Pei Han
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jia Xu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Gen Wen
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yimin Chai
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
11
|
Dallas A, Trotsyuk A, Ilves H, Bonham CA, Rodrigues M, Engel K, Barrera JA, Kosaric N, Stern-Buchbinder ZA, White A, Mandell KJ, Hammond PT, Mansbridge J, Jayasena S, Gurtner GC, Johnston BH. Acceleration of Diabetic Wound Healing with PHD2- and miR-210-Targeting Oligonucleotides. Tissue Eng Part A 2018; 25:44-54. [PMID: 29644938 DOI: 10.1089/ten.tea.2017.0484] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
In diabetes-associated chronic wounds, the normal response to hypoxia is impaired and many cellular processes involved in wound healing are hindered. Central to the hypoxia response is hypoxia-inducible factor-1α (HIF-1α), which activates multiple factors that enhance wound healing by promoting cellular motility and proliferation, new vessel formation, and re-epithelialization. Prolyl hydroxylase domain-containing protein 2 (PHD2) regulates HIF-1α activity by targeting it for degradation under normoxia. HIF-1α also upregulates microRNA miR-210, which in turn regulates proteins involved in cell cycle control, DNA repair, and mitochondrial respiration in ways that are antagonistic to wound repair. We have identified a highly potent short synthetic hairpin RNA (sshRNA) that inhibits expression of PHD2 and an antisense oligonucleotide (antimiR) that inhibits miR-210. Both oligonucleotides were chemically modified for improved biostability and to mitigate potential immunostimulatory effects. Using the sshRNA to silence PHD2 transcripts stabilizes HIF-1α and, in combination with the antimiR targeting miR-210, increases proliferation and migration of keratinocytes in vitro. To assess activity and delivery in an impaired wound healing model in diabetic mice, PHD2-targeting sshRNAs and miR-210 antimiRs both alone and in combination were formulated for local delivery to wounds using layer-by-layer (LbL) technology. LbL nanofabrication was applied to incorporate sshRNA into a thin polymer coating on a Tegaderm mesh. This coating gradually degrades under physiological conditions, releasing sshRNA and antimiR for sustained cellular uptake. Formulated treatments were applied directly to splinted full-thickness excisional wounds in db/db mice. Cellular uptake was confirmed using fluorescent sshRNA. Wounds treated with a single application of PHD2 sshRNA or antimiR-210 closed 4 days faster than untreated wounds, and wounds treated with both oligonucleotides closed on average 4.75 days faster. Markers for neovascularization and cell proliferation (CD31 and Ki67, respectively) were increased in the wound area following treatment, and vascular endothelial growth factor (VEGF) was increased in sshRNA-treated wounds. Our results suggest that silencing of PHD2 and miR-210 either together or separately by localized delivery of sshRNAs and antimiRs is a promising approach for the treatment of chronic wounds, with the potential for rapid clinical translation.
Collapse
Affiliation(s)
| | - Artem Trotsyuk
- 2 Department of Surgery, Stanford University School of Medicine, Stanford, California
| | | | - Clark A Bonham
- 2 Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Melanie Rodrigues
- 2 Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Karl Engel
- 2 Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Janos A Barrera
- 2 Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Nina Kosaric
- 2 Department of Surgery, Stanford University School of Medicine, Stanford, California
| | | | | | | | - Paula T Hammond
- 4 Koch Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | | | | | - Geoffrey C Gurtner
- 2 Department of Surgery, Stanford University School of Medicine, Stanford, California
| | | |
Collapse
|
12
|
Abstract
Diabetes is a condition that is not completely treatable but life of a diabetic patient can be smoothed by preventing or delaying the associate conditions like diabetic retinopathy, nephropathy, impaired wound healing process, etc. Apart from conventional methods to regulate diabetic condition, new techniques using siRNA have been emerged to prevent the associated conditions. This paper focuses on how siRNA used as a tool to silence the expression of genes which plays critical role in pathogenesis of these conditions. A marked improvement in wound-healing process of diabetic patients has been observed with siRNA treatment by silencing of Keap1 gene. Glucagon plays critical role in glucose homoeostasis and increases blood glucose level during hypoglycaemia. Glucose homoeostasis is impaired in diabetic patient and suppressing the expression of glucagon secretion with siRNA is used to suppress the progress of diabetes. Similarly, silencing expression of several factors has demonstrated improvement of treatment of diabetic nephropathy, retinopathy and inflammation by the use of siRNA.
Collapse
Affiliation(s)
- Pravin Shende
- a Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management , SVKM'S NMIMS , Mumbai , India
| | - Chirag Patel
- a Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management , SVKM'S NMIMS , Mumbai , India
| |
Collapse
|
13
|
Fang Y, Zhang H, Zhong Y, Ding X. Prolyl hydroxylase 2 (PHD2) inhibition protects human renal epithelial cells and mice kidney from hypoxia injury. Oncotarget 2018; 7:54317-54328. [PMID: 27527871 PMCID: PMC5342344 DOI: 10.18632/oncotarget.11104] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 07/19/2016] [Indexed: 11/25/2022] Open
Abstract
Prolyl hydroxylase domain protein 2 (PHD2) is a key oxygen sensor, setting low steady-state level of hypoxia-inducible factor-α (HIF-α). Here, we showed that treatment of cobalt chloride (CoCl2), a hypoxia mimic, in HK-2 tubular epithelial cells induced PHD2 and HIF-1/2α expression as well as cell apoptosis and autophagy activation. Three methyladenine (3-MA), the autophagy inhibitor, blocked autophagy and protected HK-2 cells from CoCl2. Significantly, siRNA knockdown of PHD2 also protected HK-2 cells from CoCl2,possibly via increasing HIF-1α expression. Reversely, HIF-1α siRNA knockdown almost abolished cytoprotection by PHD2 siRNA in CoCl2-treated HK-2 cells. In vivo, pretreatment with a PHD inhibitor L-mimosine remarkably attenuated mice renal ischemia-reperfusion injuries. Molecularly, L-mimosine inhibited apoptosis and inflammatory responses in injured mice kidneys. Together, our results suggest that PHD2 silence or inhibition protects human renal epithelial cells and mice kidney from hypoxia injuries.
Collapse
Affiliation(s)
- Yi Fang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| | - Hui Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yihong Zhong
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Kidney and Dialysis, Shanghai, China.,Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
| |
Collapse
|
14
|
Ko SH, Nauta AC, Morrison SD, Hu MS, Zimmermann AS, Chung MT, Glotzbach JP, Wong VW, Walmsley GG, Lorenz HP, Chan DA, Gurtner GC, Giaccia AJ, Longaker MT. PHD-2 Suppression in Mesenchymal Stromal Cells Enhances Wound Healing. Plast Reconstr Surg 2018; 141:55e-67e. [PMID: 29280872 PMCID: PMC5747314 DOI: 10.1097/prs.0000000000003959] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Cell therapy with mesenchymal stromal cells is a promising strategy for tissue repair. Restoration of blood flow to ischemic tissues is a key step in wound repair, and mesenchymal stromal cells have been shown to be proangiogenic. Angiogenesis is critically regulated by the hypoxia-inducible factor (HIF) superfamily, consisting of transcription factors targeted for degradation by prolyl hydroxylase domain (PHD)-2. The aim of this study was to enhance the proangiogenic capability of mesenchymal stromal cells and to use these modified cells to promote wound healing. METHODS Mesenchymal stromal cells harvested from mouse bone marrow were transduced with short hairpin RNA (shRNA) against PHD-2; control cells were transduced with scrambled shRNA (shScramble) construct. Gene expression quantification, human umbilical vein endothelial cell tube formation assays, and wound healing assays were used to assess the effect of PHD knockdown mesenchymal stromal cells on wound healing dynamics. RESULTS PHD-2 knockdown mesenchymal stromal cells overexpressed HIF-1α and multiple angiogenic factors compared to control (p < 0.05). Human umbilical vein endothelial cells treated with conditioned medium from PHD-2 knockdown mesenchymal stromal cells exhibited increased formation of capillary-like structures and enhanced migration compared with human umbilical vein endothelial cells treated with conditioned medium from shScramble-transduced mesenchymal stromal cells (p < 0.05). Wounds treated with PHD-2 knockdown mesenchymal stromal cells healed at a significantly accelerated rate compared with wounds treated with shScramble mesenchymal stromal cells (p < 0.05). Histologic studies revealed increased blood vessel density and increased cellularity in the wounds treated with PHD-2 knockdown mesenchymal stromal cells (p < 0.05). CONCLUSIONS Silencing PHD-2 in mesenchymal stromal cells augments their proangiogenic potential in wound healing therapy. This effect appears to be mediated by overexpression of HIF family transcription factors and up-regulation of multiple downstream angiogenic factors.
Collapse
Affiliation(s)
- Sae Hee Ko
- Hagey Laboratory for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA
- Division of Vascular Surgery, Department of Surgery, University of California, San Diego, CA
| | - Allison C. Nauta
- Hagey Laboratory for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Oregon Health and Sciences University, Portland, OR
| | - Shane D. Morrison
- Hagey Laboratory for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA
| | - Michael S. Hu
- Hagey Laboratory for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA
| | - Andrew S. Zimmermann
- Hagey Laboratory for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA
| | - Michael T. Chung
- Hagey Laboratory for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA
| | - Jason P. Glotzbach
- Hagey Laboratory for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA
- Division of Cardiothoracic Surgery, Department of Surgery, New York Presbyterian Hospital, New York, NY
| | - Victor W. Wong
- Hagey Laboratory for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA
| | - Graham G. Walmsley
- Hagey Laboratory for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA
| | - H. Peter Lorenz
- Hagey Laboratory for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA
| | - Denise A. Chan
- Department of Radiation Oncology, University of California, San Francisco, CA
| | - Geoffrey C. Gurtner
- Hagey Laboratory for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA
| | - Amato J. Giaccia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Michael T. Longaker
- Hagey Laboratory for Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
15
|
Stacpoole PW. Therapeutic Targeting of the Pyruvate Dehydrogenase Complex/Pyruvate Dehydrogenase Kinase (PDC/PDK) Axis in Cancer. J Natl Cancer Inst 2017; 109:3871192. [PMID: 29059435 DOI: 10.1093/jnci/djx071] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 03/27/2017] [Indexed: 02/06/2023] Open
Abstract
The mitochondrial pyruvate dehydrogenase complex (PDC) irreversibly decarboxylates pyruvate to acetyl coenzyme A, thereby linking glycolysis to the tricarboxylic acid cycle and defining a critical step in cellular bioenergetics. Inhibition of PDC activity by pyruvate dehydrogenase kinase (PDK)-mediated phosphorylation has been associated with the pathobiology of many disorders of metabolic integration, including cancer. Consequently, the PDC/PDK axis has long been a therapeutic target. The most common underlying mechanism accounting for PDC inhibition in these conditions is post-transcriptional upregulation of one or more PDK isoforms, leading to phosphorylation of the E1α subunit of PDC. Such perturbations of the PDC/PDK axis induce a "glycolytic shift," whereby affected cells favor adenosine triphosphate production by glycolysis over mitochondrial oxidative phosphorylation and cellular proliferation over cellular quiescence. Dichloroacetate is the prototypic xenobiotic inhibitor of PDK, thereby maintaining PDC in its unphosphorylated, catalytically active form. However, recent interest in the therapeutic targeting of the PDC/PDK axis for the treatment of cancer has yielded a new generation of small molecule PDK inhibitors. Ongoing investigations of the central role of PDC in cellular energy metabolism and its regulation by pharmacological effectors of PDKs promise to open multiple exciting vistas into the biochemical understanding and treatment of cancer and other diseases.
Collapse
Affiliation(s)
- Peter W Stacpoole
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, and Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL
| |
Collapse
|
16
|
Rabbani PS, Zhou A, Borab ZM, Frezzo JA, Srivastava N, More HT, Rifkin WJ, David JA, Berens SJ, Chen R, Hameedi S, Junejo MH, Kim C, Sartor RA, Liu CF, Saadeh PB, Montclare JK, Ceradini DJ. Novel lipoproteoplex delivers Keap1 siRNA based gene therapy to accelerate diabetic wound healing. Biomaterials 2017; 132:1-15. [PMID: 28391065 DOI: 10.1016/j.biomaterials.2017.04.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/29/2017] [Accepted: 04/03/2017] [Indexed: 12/20/2022]
Abstract
Therapeutics utilizing siRNA are currently limited by the availability of safe and effective delivery systems. Cutaneous diseases, specifically ones with significant genetic components are ideal candidates for topical siRNA based therapy but the anatomical structure of skin presents a considerable hurdle. Here, we optimized a novel liposome and protein hybrid nanoparticle delivery system for the topical treatment of diabetic wounds with severe oxidative stress. We utilized a cationic lipid nanoparticle (CLN) composed of 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) and the edge activator sodium cholate (NaChol), in a 6:1 ratio of DOTAP:NaChol (DNC). Addition of a cationic engineered supercharged coiled-coil protein (CSP) in a 10:1:1 ratio of DNC:CSP:siRNA produced a stable lipoproteoplex (LPP) nanoparticle, with optimal siRNA complexation, minimal cytotoxicity, and increased transfection efficacy. In a humanized murine diabetic wound healing model, our optimized LPP formulation successfully delivered siRNA targeted against Keap1, key repressor of Nrf2 which is a central regulator of redox mechanisms. Application of LPP complexing siKeap1 restored Nrf2 antioxidant function, accelerated diabetic tissue regeneration, and augmented reduction-oxidation homeostasis in the wound environment. Our topical LPP delivery system can readily be translated into clinical use for the treatment of diabetic wounds and can be extended to other cutaneous diseases with genetic components.
Collapse
Affiliation(s)
- Piul S Rabbani
- New York University School of Medicine, Hansjörg Wyss Department of Plastic and Reconstructive Surgery, 430 East 29th Street, New York, NY, 10016, United States
| | - Anna Zhou
- New York University School of Medicine, Hansjörg Wyss Department of Plastic and Reconstructive Surgery, 430 East 29th Street, New York, NY, 10016, United States
| | - Zachary M Borab
- New York University School of Medicine, Hansjörg Wyss Department of Plastic and Reconstructive Surgery, 430 East 29th Street, New York, NY, 10016, United States
| | - Joseph A Frezzo
- New York University Tandon School of Engineering, Chemical and Biomolecular Engineering Department, 6 Metrotech Center, Brooklyn, NY 11201, United States
| | - Nikita Srivastava
- New York University Tandon School of Engineering, Chemical and Biomolecular Engineering Department, 6 Metrotech Center, Brooklyn, NY 11201, United States
| | - Haresh T More
- New York University Tandon School of Engineering, Chemical and Biomolecular Engineering Department, 6 Metrotech Center, Brooklyn, NY 11201, United States
| | - William J Rifkin
- New York University School of Medicine, Hansjörg Wyss Department of Plastic and Reconstructive Surgery, 430 East 29th Street, New York, NY, 10016, United States
| | - Joshua A David
- New York University School of Medicine, Hansjörg Wyss Department of Plastic and Reconstructive Surgery, 430 East 29th Street, New York, NY, 10016, United States
| | - Samuel J Berens
- New York University Tandon School of Engineering, Chemical and Biomolecular Engineering Department, 6 Metrotech Center, Brooklyn, NY 11201, United States
| | - Raymond Chen
- New York University Tandon School of Engineering, Chemical and Biomolecular Engineering Department, 6 Metrotech Center, Brooklyn, NY 11201, United States
| | - Sophia Hameedi
- New York University School of Medicine, Hansjörg Wyss Department of Plastic and Reconstructive Surgery, 430 East 29th Street, New York, NY, 10016, United States
| | - Muhammad H Junejo
- New York University School of Medicine, Hansjörg Wyss Department of Plastic and Reconstructive Surgery, 430 East 29th Street, New York, NY, 10016, United States
| | - Camille Kim
- New York University School of Medicine, Hansjörg Wyss Department of Plastic and Reconstructive Surgery, 430 East 29th Street, New York, NY, 10016, United States
| | - Rita A Sartor
- New York University School of Medicine, Hansjörg Wyss Department of Plastic and Reconstructive Surgery, 430 East 29th Street, New York, NY, 10016, United States
| | - Che F Liu
- New York University Tandon School of Engineering, Chemical and Biomolecular Engineering Department, 6 Metrotech Center, Brooklyn, NY 11201, United States
| | - Pierre B Saadeh
- New York University School of Medicine, Hansjörg Wyss Department of Plastic and Reconstructive Surgery, 430 East 29th Street, New York, NY, 10016, United States
| | - Jin K Montclare
- New York University Tandon School of Engineering, Chemical and Biomolecular Engineering Department, 6 Metrotech Center, Brooklyn, NY 11201, United States; New York University, Chemistry Department, 100 Washington Square East, New York, NY 10003, United States
| | - Daniel J Ceradini
- New York University School of Medicine, Hansjörg Wyss Department of Plastic and Reconstructive Surgery, 430 East 29th Street, New York, NY, 10016, United States.
| |
Collapse
|
17
|
Fan X, Zhu L, Wang K, Wang B, Wu Y, Xie W, Huang C, Chan BP, Du Y. Stiffness-Controlled Thermoresponsive Hydrogels for Cell Harvesting with Sustained Mechanical Memory. Adv Healthc Mater 2017; 6. [PMID: 28105774 DOI: 10.1002/adhm.201601152] [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: 10/11/2016] [Revised: 11/23/2016] [Indexed: 01/17/2023]
Abstract
Most mechanobiological investigations focused on in situ mechanical regulation of cells on stiffness-controlled substrates with few downstream applications, as it is still challenging to harvest and expand mechanically primed cells by enzymatic digestion (e.g., trypsin) without interrupting cellular mechanical memory between passages. This study develops thermoresponsive hydrogels with controllable stiffness to generate mechanically primed cells with intact mechanical memory for augmented wound healing. No significant cellular property alteration of the fibroblasts primed on thermoresponsive hydrogels with varied stiffness has been observed through thermoresponsive harvesting. When reseeding the harvested cells for further evaluation, softer hydrogels are proven to better sustain the mechanical priming effects compared to rigid tissue culture plate, which indicates that both the stiffness-controlled substrate and thermoresponsive harvesting are required to sustain cellular mechanical memory between passages. Moreover, epigenetics analysis reveals that thermoresponsive harvesting could reduce the rearrangement and loss of chromatin proteins compared to that of trypsinization. In vivo wound healing using mechanically primed fibroblasts shows featured epithelium and sebaceous glands, which indicates augmented skin recovery compared with trypsinized fibroblasts. Thus, the thermoresponsive hydrogel-based cell harvesting system offers a powerful tool to investigate mechanobiology between cell passages and produces abundant cells with tailored mechanical priming properties for cell-based applications.
Collapse
Affiliation(s)
- Xingliang Fan
- Department of Biomedical Engineering; School of Medicine; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Tsinghua University; Beijing 100084 China
- Joint Center for Life Sciences; Tsinghua University-Peking University; Beijing 100084 China
| | - Lu Zhu
- Department of Biomedical Engineering; School of Medicine; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Tsinghua University; Beijing 100084 China
- Institute of Medical Equipment; Academy of Military Medical Sciences; Tianjin 300161 China
| | - Ke Wang
- Department of Chemistry; School of Science; Tsinghua University; Beijing 100084 China
| | - Bingjie Wang
- Department of Biomedical Engineering; School of Medicine; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Tsinghua University; Beijing 100084 China
- School of Life Science; Tsinghua University; Beijing 100084 China
| | - Yaozu Wu
- Department of Biomedical Engineering; School of Medicine; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Tsinghua University; Beijing 100084 China
| | - Wei Xie
- Joint Center for Life Sciences; Tsinghua University-Peking University; Beijing 100084 China
- School of Life Science; Tsinghua University; Beijing 100084 China
| | - Chengyu Huang
- Department of Plastic; Reconstructive and Aesthetic Surgery; Beijing Tsinghua Changgung Hospital; Tsinghua University; Beijing 102218 China
| | - Barbara Pui Chan
- Tissue Engineering Laboratory; Department of Mechanical Engineering; The University of Hong Kong; Pokfulam Road Hong Kong Special Administrative Region China
| | - Yanan Du
- Department of Biomedical Engineering; School of Medicine; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Tsinghua University; Beijing 100084 China
| |
Collapse
|
18
|
Xiao J, Chen S, Yi J, Zhang H, Ameer GA. A Cooperative Copper Metal-Organic Framework-Hydrogel System Improves Wound Healing in Diabetes. ADVANCED FUNCTIONAL MATERIALS 2017; 27:1604872. [PMID: 28729818 PMCID: PMC5513192 DOI: 10.1002/adfm.201604872] [Citation(s) in RCA: 229] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Chronic non-healing wounds remain a major clinical challenge that would benefit from the development of advanced, regenerative dressings that promote wound closure within a clinically relevant time frame. The use of copper ions has shown promise in wound healing applications possibly by promoting angiogenesis. However, reported treatments that use copper ions require multiple applications of copper salts or oxides to the wound bed, exposing the patient to potentially toxic levels of copper ions and resulting in variable outcomes. Herein we set out to assess whether copper metal organic framework nanoparticles (HKUST-1 NPs) embedded within an antioxidant thermoresponsive citrate-based hydrogel would decrease copper ion toxicity and accelerate wound healing in diabetic mice. HKUST-1 and poly-(polyethyleneglycol citrate-co-N-isopropylacrylamide) (PPCN) were synthesized and characterized. HKUST-1 NP stability in a protein solution with and without embedding them in PPCN hydrogel was determined. Copper ion release, cytotoxicity, apoptosis, and in vitro migration processes were measured. Wound closure rates and wound blood perfusion were assessed in vivo using the splinted excisional dermal wound diabetic mouse model. HKUST-1 NP disintegrated in protein solution while HKUST-1 NPs embedded in PPCN (H-HKUST-1) were protected from degradation and copper ions were slowly released. Cytotoxicity and apoptosis due to copper ion release were significantly reduced while dermal cell migration in vitro and wound closure rates in vivo were significantly enhanced. In vivo, H-HKUST-1 induced angiogenesis, collagen deposition, and re-epithelialization during wound healing in diabetic mice. These results suggest that a cooperatively stabilized, copper ion-releasing H-HKUST-1 hydrogel is a promising innovative dressing for the treatment of chronic wounds.
Collapse
Affiliation(s)
- Jisheng Xiao
- Biomedical Engineering Department, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
| | - Siyu Chen
- Biomedical Engineering Department, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
| | - Ji Yi
- Biomedical Engineering Department, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
| | - Hao Zhang
- Biomedical Engineering Department, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208
| | - Guillermo A. Ameer
- Biomedical Engineering Department, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
- Department of Surgery, Feinberg School of Medicine, Chicago, Illinois 60611, United States
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208
- Simpson Querrey Institute, Northwestern University, Chicago, IL, 60611
- Corresponding Author:
| |
Collapse
|
19
|
Manresa MC, Tambuwala MM, Radhakrishnan P, Harnoss JM, Brown E, Cavadas MA, Keogh CE, Cheong A, Barrett KE, Cummins EP, Schneider M, Taylor CT. Hydroxylase inhibition regulates inflammation-induced intestinal fibrosis through the suppression of ERK-mediated TGF-β1 signaling. [corrected]. Am J Physiol Gastrointest Liver Physiol 2016; 311:G1076-G1090. [PMID: 27789456 DOI: 10.1152/ajpgi.00229.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/09/2016] [Indexed: 01/31/2023]
Abstract
Fibrosis is a complication of chronic inflammatory disorders such as inflammatory bowel disease, a condition which has limited therapeutic options and often requires surgical intervention. Pharmacologic inhibition of oxygen-sensing prolyl hydroxylases, which confer oxygen sensitivity upon the hypoxia-inducible factor pathway, has recently been shown to have therapeutic potential in colitis, although the mechanisms involved remain unclear. Here, we investigated the impact of hydroxylase inhibition on inflammation-driven fibrosis in a murine colitis model. Mice exposed to dextran sodium sulfate, followed by a period of recovery, developed intestinal fibrosis characterized by alterations in the pattern of collagen deposition and infiltration of activated fibroblasts. Treatment with the hydroxylase inhibitor dimethyloxalylglycine ameliorated fibrosis. TGF-β1 is a key regulator of fibrosis that acts through the activation of fibroblasts. Hydroxylase inhibition reduced TGF-β1-induced expression of fibrotic markers in cultured fibroblasts, suggesting a direct role for hydroxylases in TGF-β1 signaling. This was at least in part due to inhibition of noncanonical activation of extracellular signal-regulated kinase (ERK) signaling. In summary, pharmacologic hydroxylase inhibition ameliorates intestinal fibrosis through suppression of TGF-β1-dependent ERK activation in fibroblasts. We hypothesize that in addition to previously reported immunosupressive effects, hydroxylase inhibitors independently suppress profibrotic pathways.
Collapse
Affiliation(s)
- Mario C Manresa
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland.,School of Medicine and Medical Science, Charles Institute of Dermatology, University College Dublin, Dublin, Ireland
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine, Northerm Ireland
| | - Praveen Radhakrishnan
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jonathan M Harnoss
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Eric Brown
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Miguel A Cavadas
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland.,Systems Biology Ireland, University College Dublin, Dublin, Ireland; and
| | - Ciara E Keogh
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Alex Cheong
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland.,Systems Biology Ireland, University College Dublin, Dublin, Ireland; and
| | - Kim E Barrett
- Department of Medicine and Biomedical Sciences Ph.D. Program, University of California, San Diego, School of Medicine, La Jolla, California
| | - Eoin P Cummins
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Cormac T Taylor
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland; .,Systems Biology Ireland, University College Dublin, Dublin, Ireland; and
| |
Collapse
|
20
|
Martin JR, Nelson CE, Gupta MK, Yu F, Sarett SM, Hocking KM, Pollins AC, Nanney LB, Davidson JM, Guelcher SA, Duvall CL. Local Delivery of PHD2 siRNA from ROS-Degradable Scaffolds to Promote Diabetic Wound Healing. Adv Healthc Mater 2016; 5:2751-2757. [PMID: 27717176 PMCID: PMC5152672 DOI: 10.1002/adhm.201600820] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Indexed: 12/19/2022]
Abstract
Small interfering RNA (siRNA) delivered from reactive oxygen species-degradable tissue engineering scaffolds promotes diabetic wound healing in rats. Porous poly(thioketal-urethane) scaffolds implanted in diabetic wounds locally deliver siRNA that inhibits the expression of prolyl hydroxylase domain protein 2, thereby increasing the expression of progrowth genes and increasing vasculature, proliferating cells, and tissue development in diabetic wounds.
Collapse
Affiliation(s)
- John R. Martin
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Christopher E. Nelson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Mukesh K. Gupta
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Fang Yu
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Samantha M. Sarett
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Kyle M. Hocking
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Alonda C. Pollins
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lillian B. Nanney
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffrey M. Davidson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA. Medical Research Service, Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Scott A. Guelcher
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
21
|
Iron-chelating agent desferrioxamine stimulates formation of neutrophil extracellular traps (NETs) in human blood-derived neutrophils. Biosci Rep 2016; 36:BSR20160031. [PMID: 27129288 PMCID: PMC5293572 DOI: 10.1042/bsr20160031] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 03/29/2016] [Indexed: 12/11/2022] Open
Abstract
Here we show that iron-chelating agent desferrioxamine significantly induced the formation of neutrophil extracellular traps by human blood-derived neutrophils as visualized and quantified by immunofluorescence microscopy. Further analyses characterized biochemical mechanisms associated with the NET formation by desferrioxamine. Neutrophil extracellular trap (NET) formation is a significant innate immune defense mechanism against microbial infection that complements other neutrophil functions including phagocytosis and degranulation of antimicrobial peptides. NETs are decondensed chromatin structures in which antimicrobial components (histones, antimicrobial peptides and proteases) are deployed and mediate immobilization of microbes. Here we describe an effect of iron chelation on the phenotype of NET formation. Iron-chelating agent desferrioxamine (DFO) showed a modest but significant induction of NETs by freshly isolated human neutrophils as visualized and quantified by immunocytochemistry against histone–DNA complexes. Further analyses revealed that NET induction by iron chelation required NADPH-dependent production of reactive oxygen species (ROS) as well as protease and peptidyl-arginine-deiminase 4 (PAD4) activities, three key mechanistic pathways previously linked to NET formation. Our results demonstrate that iron chelation by DFO contributes to the formation of NETs and suggest a target for pharmacological manipulation of NET activity.
Collapse
|
22
|
Abstract
Oxygen represents one of the major molecules required for the development and maintenance of life. An adequate response to hypoxia is therefore required for the functioning of the majority of living organisms and relies on the activation of the hypoxia-inducible factor (HIF) pathway. HIF prolyl hydroxylase domain-2 (PHD2) has long been recognized as the major regulator of this response, controlling a myriad of outcomes that range from cell death to proliferation. However, this enzyme has been associated with more pathways, making the role of this protein remarkably complex under distinct pathologies. While a protective role seems to exist in physiological conditions such as erythropoiesis; the picture is more complex during pathologies such as cancer. Since the regulation of this enzyme and its closest family members is currently considered as a possible therapy for various diseases, understanding the different particular roles of this protein is essential.
Collapse
Affiliation(s)
- Ana M Meneses
- Heisenberg Research Group, Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ben Wielockx
- Heisenberg Research Group, Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
23
|
Paik KJ, Maan ZN, Zielins ER, Duscher D, Whittam AJ, Morrison SD, Brett EA, Ransom RC, Hu MS, Wu JC, Gurtner GC, Longaker MT, Wan DC. Short Hairpin RNA Silencing of PHD-2 Improves Neovascularization and Functional Outcomes in Diabetic Wounds and Ischemic Limbs. PLoS One 2016; 11:e0150927. [PMID: 26967994 PMCID: PMC4788284 DOI: 10.1371/journal.pone.0150927] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 02/22/2016] [Indexed: 12/14/2022] Open
Abstract
The transcription factor hypoxia-inducible factor 1-alpha (HIF-1α) is responsible for the downstream expression of over 60 genes that regulate cell survival and metabolism in hypoxic conditions as well as those that enhance angiogenesis to alleviate hypoxia. However, under normoxic conditions, HIF-1α is hydroxylated by prolyl hydroxylase 2, and subsequently degraded, with a biological half-life of less than five minutes. Here we investigated the therapeutic potential of inhibiting HIF-1α degradation through short hairpin RNA silencing of PHD-2 in the setting of diabetic wounds and limb ischemia. Treatment of diabetic mouse fibroblasts with shPHD-2 in vitro resulted in decreased levels of PHD-2 transcript demonstrated by qRT-PCR, higher levels of HIF-1α as measured by western blot, and higher expression of the downstream angiogenic genes SDF-1 and VEGFα, as measured by qRT-PCR. In vivo, shPHD-2 accelerated healing of full thickness excisional wounds in diabetic mice compared to shScr control, (14.33 ± 0.45 days vs. 19 ± 0.33 days) and was associated with an increased vascular density. Delivery of shPHD-2 also resulted in improved perfusion of ischemic hind limbs compared to shScr, prevention of distal digit tip necrosis, and increased survival of muscle tissue. Knockdown of PHD-2 through shRNA treatment has the potential to stimulate angiogenesis through overexpression of HIF-1α and upregulation of pro-angiogenic genes downstream of HIF-1α, and may represent a viable, non-viral approach to gene therapy for ischemia related applications.
Collapse
Affiliation(s)
- Kevin J. Paik
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery Division, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Zeshaan N. Maan
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery Division, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Elizabeth R. Zielins
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery Division, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Dominik Duscher
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery Division, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Alexander J. Whittam
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery Division, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Shane D. Morrison
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery Division, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Elizabeth A. Brett
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery Division, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Ryan C. Ransom
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery Division, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Michael S. Hu
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery Division, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Joseph C. Wu
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, United States of America
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Geoffrey C. Gurtner
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery Division, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery Division, Stanford University School of Medicine, Stanford, CA, United States of America
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Derrick C. Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery Division, Stanford University School of Medicine, Stanford, CA, United States of America
- * E-mail:
| |
Collapse
|
24
|
Recent Advances in Developing Inhibitors for Hypoxia-Inducible Factor Prolyl Hydroxylases and Their Therapeutic Implications. Molecules 2015; 20:20551-68. [PMID: 26610437 PMCID: PMC6332328 DOI: 10.3390/molecules201119717] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/10/2015] [Accepted: 11/11/2015] [Indexed: 12/13/2022] Open
Abstract
Hypoxia-inducible factor (HIF) prolyl hydroxylases (PHDs) are members of the 2-oxoglutarate dependent non-heme iron dioxygenases. Due to their physiological roles in regulation of HIF-1α stability, many efforts have been focused on searching for selective PHD inhibitors to control HIF-1α levels for therapeutic applications. In this review, we first describe the structure of PHD2 as a molecular basis for structure-based drug design (SBDD) and various experimental methods developed for measuring PHD activity. We further discuss the current status of the development of PHD inhibitors enabled by combining SBDD approaches with high-throughput screening. Finally, we highlight the clinical implications of small molecule PHD inhibitors.
Collapse
|
25
|
Esfahani M, Karimi F, Afshar S, Niknazar S, Sohrabi S, Najafi R. Prolyl hydroxylase inhibitors act as agents to enhance the efficiency of cell therapy. Expert Opin Biol Ther 2015; 15:1739-55. [PMID: 26325448 DOI: 10.1517/14712598.2015.1084281] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION In stem cell-based therapy as a subtype of regenerative medicine, stem cells can be used to replace or repair injured tissue and cells in order to treat disease. Stem cells have the ability to integrate into injured areas and produce new cells via processes of proliferation and differentiation. Several studies have demonstrated that hypoxia increases self-renewal, proliferation and post-homing differentiation of stem cells through the regulation of hypoxia-inducible factor-1 (HIF-1)-mediated gene expression. Thus, pharmacological interventions including prolyl hydroxylase (PHD) inhibitors are considered as promising solutions for stem cell-based therapy. PHD inhibitors stabilize the HIF-1 and activate its pathway through preventing proteasomal degradation of HIF-1. AREAS COVERED This review focuses on the role of hypoxia, HIF-1 and especially PHD inhibitors on cell therapy. PHD structure and function are discussed as well as their inhibitors. In addition, we have investigated several preclinical studies in which PHD inhibitors improved the efficiency of cell-based therapies. EXPERT OPINION The data reviewed here suggest that PHD inhibitors are effective operators in improving stem cell therapy. However, because of some limitations, these compounds should be properly examined before clinical application.
Collapse
Affiliation(s)
- Maryam Esfahani
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Fatemeh Karimi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Saeid Afshar
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Somayeh Niknazar
- b 2 Shahid Beheshti University of Medical Science, Hearing Disorders Research Center , Tehran, the Islamic Republic of Iran
| | - Sareh Sohrabi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Rezvan Najafi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| |
Collapse
|
26
|
Deppe J, Popp T, Egea V, Steinritz D, Schmidt A, Thiermann H, Weber C, Ries C. Impairment of hypoxia-induced HIF-1α signaling in keratinocytes and fibroblasts by sulfur mustard is counteracted by a selective PHD-2 inhibitor. Arch Toxicol 2015; 90:1141-50. [PMID: 26082309 DOI: 10.1007/s00204-015-1549-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/03/2015] [Indexed: 12/31/2022]
Abstract
Skin exposure to sulfur mustard (SM) provokes long-term complications in wound healing. Similar to chronic wounds, SM-induced skin lesions are associated with low levels of oxygen in the wound tissue. Normally, skin cells respond to hypoxia by stabilization of the transcription factor hypoxia-inducible factor 1 alpha (HIF-1α). HIF-1α modulates expression of genes including VEGFA, BNIP3, and MMP2 that control processes such as angiogenesis, growth, and extracellular proteolysis essential for proper wound healing. The results of our studies revealed that exposure of primary normal human epidermal keratinocytes (NHEK) and primary normal human dermal fibroblasts (NHDF) to SM significantly impaired hypoxia-induced HIF-1α stabilization and target gene expression in these cells. Addition of a selective inhibitor of the oxygen-sensitive prolyl hydroxylase domain-containing protein 2 (PHD-2), IOX2, fully recovered HIF-1α stability, nuclear translocation, and target gene expression in NHEK and NHDF. Moreover, functional studies using a scratch wound assay demonstrated that the application of IOX2 efficiently counteracted SM-mediated deficiencies in monolayer regeneration under hypoxic conditions in NHEK and NHDF. Our findings describe a pathomechanism by which SM negatively affects hypoxia-stimulated HIF-1α signaling in keratinocytes and fibroblasts and thus possibly contributes to delayed wound healing in SM-injured patients that could be treated with PHD-2 inhibitors.
Collapse
Affiliation(s)
- Janina Deppe
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University of Munich, Pettenkoferstraße 9b, 80336, Munich, Germany
| | - Tanja Popp
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University of Munich, Pettenkoferstraße 9b, 80336, Munich, Germany
| | - Virginia Egea
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University of Munich, Pettenkoferstraße 9b, 80336, Munich, Germany
| | - Dirk Steinritz
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany.,Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | - Annette Schmidt
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany.,Molecular and Cellular Sport Medicine, German Sport University, Cologne, Germany
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University of Munich, Pettenkoferstraße 9b, 80336, Munich, Germany
| | - Christian Ries
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University of Munich, Pettenkoferstraße 9b, 80336, Munich, Germany.
| |
Collapse
|
27
|
Ruthenborg RJ, Ban JJ, Wazir A, Takeda N, Kim JW. Regulation of wound healing and fibrosis by hypoxia and hypoxia-inducible factor-1. Mol Cells 2014; 37:637-43. [PMID: 24957212 PMCID: PMC4179131 DOI: 10.14348/molcells.2014.0150] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 06/08/2014] [Indexed: 12/12/2022] Open
Abstract
Wound healing is a complex multi-step process that requires spatial and temporal orchestration of cellular and non-cellular components. Hypoxia is one of the prominent microenvironmental factors in tissue injury and wound healing. Hypoxic responses, mainly mediated by a master transcription factor of oxygen homeostasis, hypoxia-inducible factor-1 (HIF-1), have been shown to be critically involved in virtually all processes of wound healing and remodeling. Yet, mechanisms underlying hypoxic regulation of wound healing are still poorly understood. Better understanding of how the wound healing process is regulated by the hypoxic microenvironment and HIF-1 signaling pathway will provide insight into the development of a novel therapeutic strategy for impaired wound healing conditions such as diabetic wound and fibrosis. In this review, we will discuss recent studies illuminating the roles of HIF-1 in physiologic and pathologic wound repair and further, the therapeutic potentials of HIF-1 stabilization or inhibition.
Collapse
Affiliation(s)
- Robin J Ruthenborg
- Department of Molecular and Cell Biology, The University of Texas at Dallas, Richardson TX 75080, USA
| | - Jae-Jun Ban
- Department of Molecular and Cell Biology, The University of Texas at Dallas, Richardson TX 75080, USA
| | - Anum Wazir
- Department of Molecular and Cell Biology, The University of Texas at Dallas, Richardson TX 75080, USA
| | | | - Jung-whan Kim
- Department of Molecular and Cell Biology, The University of Texas at Dallas, Richardson TX 75080, USA
| |
Collapse
|