1
|
Oncel S, Basson MD. Gut homeostasis, injury, and healing: New therapeutic targets. World J Gastroenterol 2022; 28:1725-1750. [PMID: 35633906 PMCID: PMC9099196 DOI: 10.3748/wjg.v28.i17.1725] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/12/2021] [Accepted: 03/27/2022] [Indexed: 02/06/2023] Open
Abstract
The integrity of the gastrointestinal mucosa plays a crucial role in gut homeostasis, which depends upon the balance between mucosal injury by destructive factors and healing via protective factors. The persistence of noxious agents such as acid, pepsin, nonsteroidal anti-inflammatory drugs, or Helicobacter pylori breaks down the mucosal barrier and injury occurs. Depending upon the size and site of the wound, it is healed by complex and overlapping processes involving membrane resealing, cell spreading, purse-string contraction, restitution, differentiation, angiogenesis, and vasculogenesis, each modulated by extracellular regulators. Unfortunately, the gut does not always heal, leading to such pathology as peptic ulcers or inflammatory bowel disease. Currently available therapeutics such as proton pump inhibitors, histamine-2 receptor antagonists, sucralfate, 5-aminosalicylate, antibiotics, corticosteroids, and immunosuppressants all attempt to minimize or reduce injury to the gastrointestinal tract. More recent studies have focused on improving mucosal defense or directly promoting mucosal repair. Many investigations have sought to enhance mucosal defense by stimulating mucus secretion, mucosal blood flow, or tight junction function. Conversely, new attempts to directly promote mucosal repair target proteins that modulate cytoskeleton dynamics such as tubulin, talin, Ehm2, filamin-a, gelsolin, and flightless I or that proteins regulate focal adhesions dynamics such as focal adhesion kinase. This article summarizes the pathobiology of gastrointestinal mucosal healing and reviews potential new therapeutic targets.
Collapse
Affiliation(s)
- Sema Oncel
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| | - Marc D Basson
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
- Department of Surgery, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
- Department of Pathology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, United States
| |
Collapse
|
2
|
Yang GN, Strudwick XL, Bonder CS, Kopecki Z, Cowin AJ. Increased Expression of Flightless I in Cutaneous Squamous Cell Carcinoma Affects Wnt/β-Catenin Signaling Pathway. Int J Mol Sci 2021; 22:ijms222413203. [PMID: 34948000 PMCID: PMC8703548 DOI: 10.3390/ijms222413203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 11/16/2022] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) accounts for 25% of cutaneous malignancies diagnosed in Caucasian populations. Surgical removal in combination with radiation and chemotherapy are effective treatments for cSCC. Nevertheless, the aggressive metastatic forms of cSCC still have a relatively poor patient outcome. Studies have linked actin cytoskeletal dynamics and the Wnt/β-catenin signaling pathway as important modulators of cSCC pathogenesis. Previous studies have also shown that the actin-remodeling protein Flightless (Flii) is a negative regulator of cSCC. The aim of this study was to investigate if the functional effects of Flii on cSCC involve the Wnt/β-catenin signaling pathway. Flii knockdown was performed using siRNA in a human late stage aggressive metastatic cSCC cell line (MET-1) alongside analysis of Flii genetic murine models of 3-methylcholanthrene induced cSCC. Flii was increased in a MET-1 cSCC cell line and reducing Flii expression led to fewer PCNA positive cells and a concomitant reduction in cellular proliferation and symmetrical division. Knockdown of Flii led to decreased β-catenin and a decrease in the expression of the downstream effector of β-catenin signaling protein SOX9. 3-Methylcholanthrene (MCA)-induced cSCC in Flii overexpressing mice showed increased markers of cancer metastasis including talin and keratin-14 and a significant increase in SOX9 alongside a reduction in Flii associated protein (Flap-1). Taken together, this study demonstrates a role for Flii in regulating proteins involved in cSCC proliferation and tumor progression and suggests a potential role for Flii in aggressive metastatic cSCC.
Collapse
Affiliation(s)
- Gink N. Yang
- Future Industries Institute, University of South Australia, Adelaide 5095, Australia; (G.N.Y.); (X.L.S.); (Z.K.)
- Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5000, Australia;
| | - Xanthe L. Strudwick
- Future Industries Institute, University of South Australia, Adelaide 5095, Australia; (G.N.Y.); (X.L.S.); (Z.K.)
| | - Claudine S. Bonder
- Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5000, Australia;
- Adelaide Medical School, University of Adelaide, Adelaide 5000, Australia
| | - Zlatko Kopecki
- Future Industries Institute, University of South Australia, Adelaide 5095, Australia; (G.N.Y.); (X.L.S.); (Z.K.)
- Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| | - Allison J. Cowin
- Future Industries Institute, University of South Australia, Adelaide 5095, Australia; (G.N.Y.); (X.L.S.); (Z.K.)
- Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
- Correspondence: ; Tel.: +61-8-83025018
| |
Collapse
|
3
|
Seria E, Samut Tagliaferro S, Cutajar D, Galdies R, Felice A. Immunoglobulin G in Platelet-Derived Wound Healing Factors. BIOMED RESEARCH INTERNATIONAL 2021; 2021:4762657. [PMID: 33575328 PMCID: PMC7861922 DOI: 10.1155/2021/4762657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/03/2020] [Accepted: 01/13/2021] [Indexed: 11/17/2022]
Abstract
We intended to reformulate an existing platelet-derived wound healing formula to target each phase of the healing wound with the appropriate phase-specific molecules. A decreased perfusion of the skin, often associated with conditions such as thalassemia, sickle cell disease, diabetes mellitus, and chronic vascular disease, is the most common etiology of cutaneous ulcers and chronic wounds. We had previously shown that a PDWHF topically applied to a chronic nonhealing ulcer of a β-thalassemia homozygote stimulated and accelerated closure of the wound. The PDWHF was prepared from a pooled platelet concentrate of a matching blood group, consisting of a combination of platelet α-granule-derived factors. Processing of the apheresis-pooled platelets yielded various amounts of proteins (3.36 g/mL ± 0.25 (SD) (N = 10)) by the better lysis buffer method. Immunoglobulin G was found to be the most abundant α-granule-secreted protein. Equally broad quantities of the IgG (10.76 ± 12.66% (SD) (N = 10)) and IgG/albumin ratios (0.6 ± 0.4 (SD) (N = 10)) were quantified. We have developed a method using a reformulated lysis buffer followed by size exclusion chromatography and affinity chromatography to extract, identify, quantify, and purify IgG from activated platelets. IgG purification was confirmed by Western blot and flow cytometry. It was thought unlikely that the platelet IgG could be accounted for by adsorption of plasma protein, though the variable quantities could account for diversity in wound healing rates. The IgG could protect the wound even from subclinical infections and functionally advance healing. It may be useful in the management of skin ulcers in the early phase of wound healing.
Collapse
Affiliation(s)
- Elisa Seria
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, Centre of Molecular Medicine and Biobanking, University of Malta and Division of Pathology, Mater Dei Hospital, Malta MSD2080
| | - Sarah Samut Tagliaferro
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, Centre of Molecular Medicine and Biobanking, University of Malta and Division of Pathology, Mater Dei Hospital, Malta MSD2080
| | - Doreen Cutajar
- Department of Surgery, Faculty of Medicine and Surgery, University of Malta Medical School and Mater Dei Hospital, Malta MSD2080
| | - Ruth Galdies
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, Centre of Molecular Medicine and Biobanking, University of Malta and Division of Pathology, Mater Dei Hospital, Malta MSD2080
| | - Alex Felice
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, Centre of Molecular Medicine and Biobanking, University of Malta and Division of Pathology, Mater Dei Hospital, Malta MSD2080
| |
Collapse
|
4
|
Strudwick XL, Cowin AJ. Multifunctional Roles of the Actin-Binding Protein Flightless I in Inflammation, Cancer and Wound Healing. Front Cell Dev Biol 2020; 8:603508. [PMID: 33330501 PMCID: PMC7732498 DOI: 10.3389/fcell.2020.603508] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/30/2020] [Indexed: 11/20/2022] Open
Abstract
Flightless I is an actin-binding member of the gelsolin family of actin-remodeling proteins that inhibits actin polymerization but does not possess actin severing ability. Flightless I functions as a regulator of many cellular processes including proliferation, differentiation, apoptosis, and migration all of which are important for many physiological processes including wound repair, cancer progression and inflammation. More than simply facilitating cytoskeletal rearrangements, Flightless I has other important roles in the regulation of gene transcription within the nucleus where it interacts with nuclear hormone receptors to modulate cellular activities. In conjunction with key binding partners Leucine rich repeat in the Flightless I interaction proteins (LRRFIP)1/2, Flightless I acts both synergistically and competitively to regulate a wide range of cellular signaling including interacting with two of the most important inflammatory pathways, the NLRP3 inflammasome and the MyD88-TLR4 pathways. In this review we outline the current knowledge about this important cytoskeletal protein and describe its many functions across a range of health conditions and pathologies. We provide perspectives for future development of Flightless I as a potential target for clinical translation and insights into potential therapeutic approaches to manipulate Flightless I functions.
Collapse
Affiliation(s)
- Xanthe L Strudwick
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
| | - Allison J Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
| |
Collapse
|
5
|
Strudwick XL, Adams DH, Pyne NT, Samuel MS, Murray RZ, Cowin AJ. Systemic Delivery of Anti-Integrin αL Antibodies Reduces Early Macrophage Recruitment, Inflammation, and Scar Formation in Murine Burn Wounds. Adv Wound Care (New Rochelle) 2020; 9:637-648. [PMID: 33124967 PMCID: PMC7698651 DOI: 10.1089/wound.2019.1035] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 11/17/2019] [Indexed: 12/30/2022] Open
Abstract
Objective: Increased macrophage recruitment in the early stages of wound healing leads to an excessive inflammatory response associated with elevated fibrosis and scarring. This recruitment relies upon integrins on the surface of monocytes that regulate their migration and extravasation from the circulation into the wound site, where they differentiate into macrophages. The aim of this study was to determine if inhibiting monocyte extravasation from the circulation into burns would reduce macrophages numbers in burns and lead to reduced inflammation and scar formation. Approach: Scald burns were created on mice and treated with integrin alpha L (αL) function blocking antibody via intravenous delivery day 1 after injury. The effect of inhibiting macrophage recruitment into the burn was assessed using macro- and microscopic wound parameters as well as immunohistochemistry for inflammatory cell markers, cytokines, and collagen deposition. Results: Burn wound-associated macrophages were reduced by 54.7% at day 3 following treatment with integrin αL antibody, with levels returning to normal by day 7. This reduction in macrophages led to a concomitant reduction in inflammatory mediators, including tumor necrosis factor-alpha (TNFα) and Il-10 as well as a reduction in proscarring transforming growth factor beta 1 (TGFβ1). This reduced inflammatory response was also associated with less alpha smooth muscle actin (αSMA) expression and an overall trend toward reduced scar formation with a lower collagen I/III ratio. Innovation: Treatment of burns with integrin αL function blocking antibodies reduces inflammation in burn wounds. Conclusion: These results suggest that reducing macrophage infiltration into burn wounds may lead to a reduced early inflammatory response and less scar formation following burn injury.
Collapse
Affiliation(s)
- Xanthe L. Strudwick
- Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Damian H. Adams
- Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Natasha T. Pyne
- Centre for Cancer Biology, University of South Australia, Adelaide, South Australia, Australia
| | - Michael S. Samuel
- Centre for Cancer Biology, University of South Australia, Adelaide, South Australia, Australia
- Faculty of Health and Medical Sciences, School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Rachael Z. Murray
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Allison J. Cowin
- Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
6
|
Jackson JE, Kopecki Z, Anderson PJ, Cowin AJ. Increasing the level of cytoskeletal protein Flightless I reduces adhesion formation in a murine digital flexor tendon model. J Orthop Surg Res 2020; 15:362. [PMID: 32854733 PMCID: PMC7450967 DOI: 10.1186/s13018-020-01889-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/13/2020] [Indexed: 12/29/2022] Open
Abstract
Background Surgical repair of tendons is common, but function is often limited due to the formation of flexor tendon adhesions which reduce the mobility and use of the affected digit and hand. The severity of adhesion formation is dependent on numerous cellular processes many of which involve the actin cytoskeleton. Flightless I (Flii) is a highly conserved cytoskeletal protein, which has previously been identified as a potential target for improved healing of tendon injuries. Using human in vitro cell studies in conjunction with a murine model of partial laceration of the digital flexor tendon, we investigated the effect of modulating Flii levels on tenocyte function and formation of adhesions. Methods Human tenocyte proliferation and migration was determined using WST-1 and scratch wound assays following Flii knockdown by siRNA in vitro. Additionally, mice with normal and increased levels of Flii were subjected to a partial laceration of the digital flexor tendon in conjunction with a full tenotomy to immobilise the paw. Resulting adhesions were assessed using histology and immunohistochemistry for collagen I, III, TGF-β1and -β3 Results Flii knockdown significantly reduced human tenocyte proliferation and migration in vitro. Increasing the expression of Flii significantly reduced digital tendon adhesion formation in vivo which was confirmed through significantly smaller adhesion scores based on collagen fibre orientation, thickness, proximity to other fibres and crimping. Reduced adhesion formation was accompanied with significantly decreased deposition of type I collagen and increased expression of TGF-β1 in vivo. Conclusions These findings suggest that increasing the level of Flii in an injured tendon may be beneficial for decreasing tendon adhesion formation.
Collapse
Affiliation(s)
- Jessica E Jackson
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Zlatko Kopecki
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Peter J Anderson
- Faculty of Medicine and Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Allison J Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia.
| |
Collapse
|
7
|
Thomas HM, Ahangar P, Hofma BR, Strudwick XL, Fitridge R, Mills SJ, Cowin AJ. Attenuation of Flightless I Increases Human Pericyte Proliferation, Migration and Angiogenic Functions and Improves Healing in Murine Diabetic Wounds. Int J Mol Sci 2020; 21:ijms21165599. [PMID: 32764293 PMCID: PMC7460558 DOI: 10.3390/ijms21165599] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 12/14/2022] Open
Abstract
Pericytes are peri-vascular mural cells which have an important role in the homeostatic regulation of inflammatory and angiogenic processes. Flightless I (Flii) is a cytoskeletal protein involved in regulating cellular functions, but its involvement in pericyte activities during wound healing is unknown. Exacerbated inflammation and reduced angiogenesis are hallmarks of impaired diabetic healing responses, and strategies aimed at regulating these processes are vital for improving healing outcomes. To determine the effect of altering Flii expression on pericyte function, in vitro and in vivo studies were performed to assess the effect on healing, inflammation and angiogenesis in diabetic wounds. Here, we demonstrated that human diabetic wounds display upregulated expression of the Flii protein in conjunction with a depletion in the number of platelet derived growth factor receptor β (PDGFRβ) +/ neural glial antigen 2 (NG2) + pericytes present in the dermis. Human pericytes were found to be positive for Flii and attenuating its expression in vitro through siRNA knockdown led to enhanced proliferation, migration and angiogenic functions. Genetic knockdown of Flii in a streptozotocin-induced murine model of diabetes led to increased numbers of pericytes within the wound. This was associated with dampened inflammation, an increased rate of angiogenic repair and improved wound healing. Our findings show that Flii expression directly impacts pericyte functions, including proliferation, motility and angiogenic responses. This suggests that Flii regulation of pericyte function may be in part responsible for the changes in pericyte-related processes observed in diabetic wounds.
Collapse
Affiliation(s)
- Hannah M Thomas
- Future Industries Institute, University of South Australia, Adelaide 5000, Australia; (H.M.T.); (P.A.); (B.R.H.); (X.L.S.); (S.J.M.)
- Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
- Cell Therapy Manufacturing Cooperative Research Centre, Adelaide 5000, Australia
| | - Parinaz Ahangar
- Future Industries Institute, University of South Australia, Adelaide 5000, Australia; (H.M.T.); (P.A.); (B.R.H.); (X.L.S.); (S.J.M.)
- Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
- Cell Therapy Manufacturing Cooperative Research Centre, Adelaide 5000, Australia
| | - Benjamin R Hofma
- Future Industries Institute, University of South Australia, Adelaide 5000, Australia; (H.M.T.); (P.A.); (B.R.H.); (X.L.S.); (S.J.M.)
- Cell Therapy Manufacturing Cooperative Research Centre, Adelaide 5000, Australia
| | - Xanthe L Strudwick
- Future Industries Institute, University of South Australia, Adelaide 5000, Australia; (H.M.T.); (P.A.); (B.R.H.); (X.L.S.); (S.J.M.)
| | - Robert Fitridge
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, Australia;
| | - Stuart J Mills
- Future Industries Institute, University of South Australia, Adelaide 5000, Australia; (H.M.T.); (P.A.); (B.R.H.); (X.L.S.); (S.J.M.)
- Cell Therapy Manufacturing Cooperative Research Centre, Adelaide 5000, Australia
| | - Allison J Cowin
- Future Industries Institute, University of South Australia, Adelaide 5000, Australia; (H.M.T.); (P.A.); (B.R.H.); (X.L.S.); (S.J.M.)
- Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
- Correspondence: ; Tel.: +61-883-025-018
| |
Collapse
|
8
|
Rajeev G, Melville E, Cowin AJ, Prieto-Simon B, Voelcker NH. Porous Alumina Membrane-Based Electrochemical Biosensor for Protein Biomarker Detection in Chronic Wounds. Front Chem 2020; 8:155. [PMID: 32211379 PMCID: PMC7067747 DOI: 10.3389/fchem.2020.00155] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 02/20/2020] [Indexed: 12/16/2022] Open
Abstract
A label-free electrochemical detection platform for the sensitive and rapid detection of Flightless I (Flii) protein, a biomarker of wound chronicity, has been developed using nanoporous anodic alumina (NAA) membranes modified with Flii antibody recognition sites. The electrochemical detection is based on the nanochannel blockage experienced upon Flii capture by immobilized antibodies within the nanochannels. This capture impedes the diffusion of redox species [[Fe(CN)6]4-/3-] toward a gold electrode attached at the backside of the modified NAA membrane. Partial blockage causes a decrease in the oxidation current of the redox species at the electrode surface which is used as an analytical signal by the reported biosensor. The resulting biosensing system allows detection of Flii at the levels found in wounds. Two types of assays were tested, sandwich and direct, showing <3 and 2 h analysis time, respectively, a significant reduction in time from the nearly 48 h required for the conventional Western blot assay. Slightly higher sensitivity values were observed for the sandwich-based strategy. With faster analysis, lack of matrix effects, robustness, ease of use and cost-effectiveness, the developed sensing platform has the potential to be translated into a point-of-care (POC) device for chronic wound management and as a simple alternative characterization tool in Flii research.
Collapse
Affiliation(s)
- Gayathri Rajeev
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
| | - Elizabeth Melville
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
| | - Allison J Cowin
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
| | - Beatriz Prieto-Simon
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia.,Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Department of Electronic Engineering, Universitat Rovira i Virgili, Tarragona, Spain
| | - Nicolas H Voelcker
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia.,Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Clayton, VIC, Australia.,Melbourne Centre for Nanofabrication, Clayton, VIC, Australia.,Materials Science and Engineering, Monash University, Clayton, VIC, Australia
| |
Collapse
|
9
|
Kopecki Z, Yang G, Treloar S, Mashtoub S, Howarth GS, Cummins AG, Cowin AJ. Flightless I exacerbation of inflammatory responses contributes to increased colonic damage in a mouse model of dextran sulphate sodium-induced ulcerative colitis. Sci Rep 2019; 9:12792. [PMID: 31488864 PMCID: PMC6728368 DOI: 10.1038/s41598-019-49129-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/15/2019] [Indexed: 12/13/2022] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by cytokine driven inflammation that disrupts the mucosa and impedes intestinal structure and functions. Flightless I (Flii) is an immuno-modulatory protein is a member of the gelsolin family of actin-remodelling proteins that regulates cellular and inflammatory processes critical in tissue repair. Here we investigated its involvement in UC and show that Flii is significantly elevated in colonic tissues of patients with inflammatory bowel disease. Using an acute murine model of colitis, we characterised the contribution of Flii to UC using mice with low (Flii+/-), normal (Flii+/+) and high Flii (FliiTg/Tg). High levels of Flii resulted in significantly elevated disease severity index scores, increased rectal bleeding and degree of colon shortening whereas, low Flii expression decreased disease severity, reduced tissue inflammation and improved clinical indicators of UC. Mice with high levels of Flii had significantly increased histological disease severity and elevated mucosal damage with significantly increased inflammatory cell infiltrate and significantly higher levels of TNF-α, IFN-γ, IL-5 and IL-13 pro-inflammatory cytokines. Additionally, Flii overexpression resulted in decreased β-catenin levels, inhibited Wnt/β-catenin signalling and impaired regeneration of colonic crypts. These studies suggest that high levels of Flii, as is observed in patients with UC, may adversely affect mucosal healing via mechanisms involving Th1 and Th2 mediated tissue inflammation and Wnt/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Z Kopecki
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide, South Australia, Australia.
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia.
| | - G Yang
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide, South Australia, Australia
| | - S Treloar
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, Adelaide, South Australia, Australia
| | - S Mashtoub
- Department of Gastroenterology, Women's and Children's Hospital, North Adelaide, South Australia, Australia
- Discipline of Physiology, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - G S Howarth
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - A G Cummins
- Department of Gastroenterology and Hepatology, The Queen Elizabeth Hospital, Woodville South, Adelaide, South Australia, Australia
| | - A J Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide, South Australia, Australia
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
10
|
Rajeev G, Cowin AJ, Voelcker NH, Prieto Simon B. Magnetic Nanoparticles Enhance Pore Blockage-Based Electrochemical Detection of a Wound Biomarker. Front Chem 2019; 7:438. [PMID: 31245362 PMCID: PMC6582131 DOI: 10.3389/fchem.2019.00438] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 05/28/2019] [Indexed: 11/29/2022] Open
Abstract
A novel pore blockage-based electrochemical immunosensor based on the combination of 100 nm-magnetic nanoparticles (MNPs), as signal enhancers, and 200 nm-pore diameter nanoporous anodic alumina (NAA) membranes, as sensing platform, is reported. A peptide conjugate mimicking flightless I (Flii), a wound healing biomarker, was chosen as target analyte. The sensing platform consists of an anti-Flii antibody (Ab1)-modified NAA membrane attached onto a gold electrode. Anti-KLH antibody (Ab2)-modified MNPs (MNP-Ab2) were used to selectively capture the Flii peptide conjugate in solution. Sensing was based on pore blockage of the Ab1-modified NAA membrane caused upon specific binding of the MNP-Ab2-analyte complex. The degree of pore blockage, and thus the concentration of the Flii peptide conjugate in the sample, was measured as a reduction in the oxidation current of a redox species ([Fe(CN)6]4-) added in solution. We demonstrated that pore blockage is drastically enhanced by applying an external magnetic field at the membrane backside to facilitate access of the MNP-Ab2-analyte complex into the pores, and thus ensure its availability to bind to the Ab1-modified NAA membrane. Combining the pore blockage-based electrochemical magnetoimmunosensor with an externally applied magnetic field, a limit of detection (LOD) of 0.5 ng/ml of Flii peptide conjugate was achieved, while sensing in the absence of magnetic field could only attain a LOD of 1.2 μg/ml. The developed sensing strategy is envisaged as a powerful solution for the ultra-sensitive detection of an analyte of interest present in a complex matrix.
Collapse
Affiliation(s)
- Gayathri Rajeev
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
- Faculty of Science, Institute for Biomedical Materials and Devices, University of Technology, Sydney, NSW, Australia
| | - Allison J. Cowin
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
| | - Nicolas H. Voelcker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, Australia
- Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, Australia
| | - Beatriz Prieto Simon
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, Australia
- Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia
| |
Collapse
|
11
|
Takimoto M. Multidisciplinary Roles of LRRFIP1/GCF2 in Human Biological Systems and Diseases. Cells 2019; 8:cells8020108. [PMID: 30709060 PMCID: PMC6406849 DOI: 10.3390/cells8020108] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/21/2019] [Accepted: 01/27/2019] [Indexed: 01/28/2023] Open
Abstract
Leucine Rich Repeat of Flightless-1 Interacting Protein 1/GC-binding factor 2 (LRRFIP1/GCF2) cDNA was cloned for a transcriptional repressor GCF2, which bound sequence-specifically to a GC-rich element of epidermal growth factor receptor (EGFR) gene and repressed its promotor. LRRFIP1/GCF2 was also cloned as a double stranded RNA (dsRNA)-binding protein to trans-activation responsive region (TAR) RNA of Human Immunodeficiency Virus-1 (HIV-1), termed as TAR RNA interacting protein (TRIP), and as a binding protein to the Leucine Rich Repeat (LRR) of Flightless-1(Fli-1), termed as Flightless-1 LRR associated protein 1 (FLAP1) and LRR domain of Flightless-1 interacting Protein 1 (LRRFIP1). Subsequent functional studies have revealed that LRRFIP1/GCF2 played multiple roles in the regulation of diverse biological systems and processes, such as in immune response to microorganisms and auto-immunity, remodeling of cytoskeletal system, signal transduction pathways, and transcriptional regulations of genes. Dysregulations of LRRFIP1/GCF2 have been implicated in the causes of several experimental and clinico-pathological states and the responses to them, such as autoimmune diseases, excitotoxicity after stroke, thrombosis formation, inflammation and obesity, the wound healing process, and in cancers. LRRFIP1/GCF2 is a bioregulator in multidisciplinary systems of the human body and its dysregulation can cause diverse human diseases.
Collapse
Affiliation(s)
- Masato Takimoto
- Institute for Genetic Medicine, Hokkaido University, Hokkaido 060-0815, Japan.
| |
Collapse
|
12
|
Kopecki Z, Stevens NE, Yang GN, Melville E, Cowin AJ. Recombinant Leucine-Rich Repeat Flightless-Interacting Protein-1 Improves Healing of Acute Wounds through Its Effects on Proliferation Inflammation and Collagen Deposition. Int J Mol Sci 2018; 19:ijms19072014. [PMID: 29996558 PMCID: PMC6073877 DOI: 10.3390/ijms19072014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/04/2018] [Accepted: 07/07/2018] [Indexed: 02/07/2023] Open
Abstract
Wound healing is an increasing clinical problem involving substantial morbidity, mortality, and rising health care costs. Leucine-rich repeat flightless-interacting protein-1 (LRRFIP-1) regulates toll-like receptor (TLR)-mediated inflammation, suggesting a potential role in the healing of wounds. We sought to determine the role of LRRFIP-1 in wound repair and whether the exogenous addition of recombinant LRRFIP-1 (rLRRFIP-1) affected healing responses. Using a model of full-thickness incisional acute wounds in BALB/c mice, we investigated the effect of wounding on LRRFIP-1 expression. The effect of rLRRFIP-1 on cellular proliferation, inflammation, and collagen deposition was also investigated. LRRFIP-1 was upregulated in response to wounding, was found to directly associate with flightless I (Flii), and significantly increased cellular proliferation both in vitro and in vivo. rLRRFIP-1 reduced Flii expression in wounds in vivo and resulted in significantly improved healing with a concurrent dampening of TLR4-mediated inflammation and improved collagen deposition. Additionally, decreased levels of TGF-β1 and increased levels of TGF-β3 were observed in rLRRFIP-1-treated wounds suggesting a possible antiscarring effect of rLRRFIP-1. Further studies are required to elucidate if the mechanisms behind LRRFIP-1 action in wound repair are independent of Flii. However, these results identify rLRRFIP-1 as a possible treatment modality for improved healing of acute wounds.
Collapse
Affiliation(s)
- Zlatko Kopecki
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide SA 5095, Australia.
| | - Natalie E Stevens
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide SA 5095, Australia.
| | - Gink N Yang
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide SA 5095, Australia.
| | - Elizabeth Melville
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide SA 5095, Australia.
| | - Allison J Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide SA 5095, Australia.
| |
Collapse
|
13
|
Xu L, Carrer A, Zonta F, Qu Z, Ma P, Li S, Ceriani F, Buratto D, Crispino G, Zorzi V, Ziraldo G, Bruno F, Nardin C, Peres C, Mazzarda F, Salvatore AM, Raspa M, Scavizzi F, Chu Y, Xie S, Yang X, Liao J, Liu X, Wang W, Wang S, Yang G, Lerner RA, Mammano F. Design and Characterization of a Human Monoclonal Antibody that Modulates Mutant Connexin 26 Hemichannels Implicated in Deafness and Skin Disorders. Front Mol Neurosci 2017; 10:298. [PMID: 29018324 PMCID: PMC5615210 DOI: 10.3389/fnmol.2017.00298] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/05/2017] [Indexed: 12/21/2022] Open
Abstract
Background: Mutations leading to changes in properties, regulation, or expression of connexin-made channels have been implicated in 28 distinct human hereditary diseases. Eight of these result from variants of connexin 26 (Cx26), a protein critically involved in cell-cell signaling in the inner ear and skin. Lack of non-toxic drugs with defined mechanisms of action poses a serious obstacle to therapeutic interventions for diseases caused by mutant connexins. In particular, molecules that specifically modulate connexin hemichannel function without affecting gap junction channels are considered of primary importance for the study of connexin hemichannel role in physiological as well as pathological conditions. Monoclonal antibodies developed in the last three decades have become the most important class of therapeutic biologicals. Recombinant methods permit rapid selection and improvement of monoclonal antibodies from libraries with large diversity. Methods: By screening a combinatorial library of human single-chain fragment variable (scFv) antibodies expressed in phage, we identified a candidate that binds an extracellular epitope of Cx26. We characterized antibody action using a variety of biochemical and biophysical assays in HeLa cells, organotypic cultures of mouse cochlea and human keratinocyte-derived cells. Results: We determined that the antibody is a remarkably efficient, non-toxic, and completely reversible inhibitor of hemichannels formed by connexin 26 and does not affect direct cell-cell communication via gap junction channels. Importantly, we also demonstrate that the antibody efficiently inhibits hyperative mutant Cx26 hemichannels implicated in autosomal dominant non-syndromic hearing impairment accompanied by keratitis and hystrix-like ichthyosis-deafness (KID/HID) syndrome. We solved the crystal structure of the antibody, identified residues that are critical for binding and used molecular dynamics to uncover its mechanism of action. Conclusions: Although further studies will be necessary to validate the effect of the antibody in vivo, the methodology described here can be extended to select antibodies against hemichannels composed by other connexin isoforms and, consequently, to target other pathologies associated with hyperactive hemichannels. Our study highlights the potential of this approach and identifies connexins as therapeutic targets addressable by screening phage display libraries expressing human randomized antibodies.
Collapse
Affiliation(s)
- Liang Xu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institute for Biological Sciences, Chinese Academy of SciencesShanghai, China
| | - Andrea Carrer
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy
| | - Francesco Zonta
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China.,CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy
| | - Zhihu Qu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Peixiang Ma
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Sheng Li
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Federico Ceriani
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy
| | - Damiano Buratto
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy
| | - Giulia Crispino
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy.,Venetian Institute of Molecular MedicinePadova, Italy
| | - Veronica Zorzi
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Institute of Otolaryngology, Catholic University School of MedicineRome, Italy
| | - Gaia Ziraldo
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy.,Institute of Otolaryngology, Catholic University School of MedicineRome, Italy
| | - Francesca Bruno
- Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy.,Venetian Institute of Molecular MedicinePadova, Italy
| | - Chiara Nardin
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Science, Roma Tre UniversityRome, Italy
| | - Chiara Peres
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy
| | - Flavia Mazzarda
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Science, Roma Tre UniversityRome, Italy
| | - Anna M Salvatore
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy
| | - Marcello Raspa
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy
| | | | - Youjun Chu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Sichun Xie
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Xuemei Yang
- School of Life Science and Technology, Shanghai Tech UniversityShanghai, China
| | - Jun Liao
- School of Life Science and Technology, Shanghai Tech UniversityShanghai, China
| | - Xiao Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institute for Biological Sciences, Chinese Academy of SciencesShanghai, China.,University of Chinese Academy of SciencesBeijing, China
| | - Wei Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Shanshan Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Guang Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Richard A Lerner
- Department of Cell and Molecular Biology, The Scripps Research InstituteLa Jolla, CA, United States
| | - Fabio Mammano
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China.,CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy.,Venetian Institute of Molecular MedicinePadova, Italy
| |
Collapse
|
14
|
Kopecki Z, Yang GN, Jackson JE, Melville EL, Calley MP, Murrell DF, Darby IA, O'Toole EA, Samuel MS, Cowin AJ. Cytoskeletal protein Flightless I inhibits apoptosis, enhances tumor cell invasion and promotes cutaneous squamous cell carcinoma progression. Oncotarget 2017; 6:36426-40. [PMID: 26497552 PMCID: PMC4742187 DOI: 10.18632/oncotarget.5536] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 10/09/2015] [Indexed: 01/08/2023] Open
Abstract
Flightless I (Flii) is an actin remodeling protein that affects cellular processes including adhesion, proliferation and migration. In order to determine the role of Flii during carcinogenesis, squamous cell carcinomas (SCCs) were induced in Flii heterozygous (Flii+/-), wild-type and Flii overexpressing (FliiTg/Tg) mice by intradermal injection of 3-methylcholanthrene (MCA). Flii levels were further assessed in biopsies from human SCCs and the human SCC cell line (MET-1) was used to determine the effect of Flii on cellular invasion. Flii was highly expressed in human SCC biopsies particularly by the invading cells at the tumor edge. FliiTg/Tg mice developed large, aggressive SCCs in response to MCA. In contrast Flii+/- mice had significantly smaller tumors that were less invasive. Intradermal injection of Flii neutralizing antibodies during SCC initiation and progression significantly reduced the size of the tumors and, in vitro, decreased cellular sphere formation and invasion. Analysis of the tumors from the Flii overexpressing mice showed reduced caspase I and annexin V expression suggesting Flii may negatively regulate apoptosis within these tumors. These studies therefore suggest that Flii enhances SCC tumor progression by decreasing apoptosis and enhancing tumor cell invasion. Targeting Flii may be a potential strategy for reducing the severity of SCCs.
Collapse
Affiliation(s)
- Zlatko Kopecki
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Gink N Yang
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Jessica E Jackson
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Elizabeth L Melville
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Matthew P Calley
- Centre for Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Dedee F Murrell
- Department of Dermatology, St. George Hospital and University of New South Wales, Sydney, New South Wales, Australia
| | - Ian A Darby
- School of Medical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Edel A O'Toole
- Centre for Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Michael S Samuel
- Centre for Cancer Biology, an alliance between SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - Allison J Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
15
|
Turner CT, Hasanzadeh Kafshgari M, Melville E, Delalat B, Harding F, Mäkilä E, Salonen JJ, Cowin AJ, Voelcker NH. Delivery of Flightless I siRNA from Porous Silicon Nanoparticles Improves Wound Healing in Mice. ACS Biomater Sci Eng 2016; 2:2339-2346. [DOI: 10.1021/acsbiomaterials.6b00550] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Christopher T. Turner
- Regenerative Medicine, Future
Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Morteza Hasanzadeh Kafshgari
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Future Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Elizabeth Melville
- Regenerative Medicine, Future
Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Bahman Delalat
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Future Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Francis Harding
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Future Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Ermei Mäkilä
- Department
of Physics and Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Jarno J. Salonen
- Department
of Physics and Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Allison J. Cowin
- Regenerative Medicine, Future
Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Nicolas H. Voelcker
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Future Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| |
Collapse
|
16
|
Kopecki Z, Ludwig RJ, Cowin AJ. Cytoskeletal Regulation of Inflammation and Its Impact on Skin Blistering Disease Epidermolysis Bullosa Acquisita. Int J Mol Sci 2016; 17:ijms17071116. [PMID: 27420054 PMCID: PMC4964491 DOI: 10.3390/ijms17071116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 06/30/2016] [Accepted: 07/07/2016] [Indexed: 01/10/2023] Open
Abstract
Actin remodelling proteins regulate cytoskeletal cell responses and are important in both innate and adaptive immunity. These responses play a major role in providing a fine balance in a cascade of biological events that results in either protective acute inflammation or chronic inflammation that leads to a host of diseases including autoimmune inflammation mediated epidermolysis bullosa acquisita (EBA). This review describes the role of the actin cytoskeleton and in particular the actin remodelling protein called Flightless I (Flii) in regulating cellular inflammatory responses and its subsequent effect on the autoimmune skin blistering disease EBA. It also outlines the potential of an antibody based therapy for decreasing Flii expression in vivo to ameliorate the symptoms associated with EBA.
Collapse
Affiliation(s)
- Zlatko Kopecki
- Future Industries Institute, Regenerative Medicine, University of South Australia, Mawson Lakes 5095, Adelaide, Australia.
| | - Ralf J Ludwig
- Institute of Experimental Dermatology, University of Lubeck, Lubeck 23562, Germany.
| | - Allison J Cowin
- Future Industries Institute, Regenerative Medicine, University of South Australia, Mawson Lakes 5095, Adelaide, Australia.
| |
Collapse
|
17
|
Ruzehaji N, Kopecki Z, Melville E, Appleby SL, Bonder CS, Arkell RM, Fitridge R, Cowin AJ. Attenuation of flightless I improves wound healing and enhances angiogenesis in a murine model of type 1 diabetes. Diabetologia 2014; 57:402-12. [PMID: 24292564 DOI: 10.1007/s00125-013-3107-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 10/18/2013] [Indexed: 10/26/2022]
Abstract
AIMS/HYPOTHESIS Skin lesions and ulcerations are severe complications of diabetes that often result in leg amputations. In this study we investigated the function of the cytoskeletal protein flightless I (FLII) in diabetic wound healing. We hypothesised that overexpression of FLII would have a negative effect on diabetic wound closure and modulation of this protein using specific FLII-neutralising antibodies (FnAb) would enhance cellular proliferation, migration and angiogenesis within the diabetic wound. METHODS Using a streptozotocin-induced model of diabetes we investigated the effect of altered FLII levels through Flii genetic knockdown, overexpression or treatment with FnAb on wound healing. Diabetic wounds were assessed using histology, immunohistochemistry and biochemical analysis. In vitro and in vivo assays of angiogenesis were used to assess the angiogenic response. RESULTS FLII levels were elevated in the wounds of both diabetic mice and humans. Reduction in the level of FLII improved healing of murine diabetic wounds and promoted a robust pro-angiogenic response with significantly elevated von Willebrand factor (vWF) and vascular endothelial growth factor (VEGF)-positive endothelial cell infiltration. Diabetic mouse wounds treated intradermally with FnAb showed improved healing and a significantly increased rate of re-epithelialisation. FnAb improved the angiogenic response through enhanced formation of capillary tubes and functional neovasculature. Reducing the level of FLII led to increased numbers of mature blood vessels, increased recruitment of smooth muscle actin-α-positive cells and improved tight junction formation. CONCLUSIONS/INTERPRETATION Reducing the level of FLII in a wound may be a potential therapeutic approach for the treatment of diabetic foot ulcers.
Collapse
Affiliation(s)
- Nadira Ruzehaji
- Women's and Children's Health Research Institute, Adelaide, SA, Australia
| | | | | | | | | | | | | | | |
Collapse
|
18
|
The influence of Flightless I on Toll-like-receptor-mediated inflammation in a murine model of diabetic wound healing. BIOMED RESEARCH INTERNATIONAL 2013; 2013:389792. [PMID: 23555084 PMCID: PMC3595111 DOI: 10.1155/2013/389792] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 01/07/2013] [Accepted: 01/07/2013] [Indexed: 02/06/2023]
Abstract
Impaired wound healing and ulceration represent a serious complication of both type 1 and type 2 diabetes. Cytoskeletal protein Flightless I (Flii) is an important inhibitor of wound repair, and reduced Flii gene expression in fibroblasts increased migration, proliferation, and adhesion. As such it has the ability to influence all phases of wound healing including inflammation, remodelling and angiogenesis. Flii has the potential to modulate inflammation through its interaction with MyD88 which it an adaptor protein for TLR4. To assess the effect of Flii on the inflammatory response of diabetic wounds, we used a murine model of streptozocin-induced diabetes and Flii genetic mice. Increased levels of Flii were detected in Flii transgenic murine wounds resulting in impaired healing which was exacerbated when diabetes was induced. When Flii levels were reduced in diabetic wounds of Flii-deficient mice, healing was improved and decreased levels of TLR4 were observed. In contrast, increasing the level of Flii in diabetic mouse wounds led to increased TLR4 and NF- κ B production. Treatment of murine diabetic wounds with neutralising antibodies to Flii led to an improvement in healing with decreased expression of TLR4. Decreasing the level of Flii in diabetic wounds may therefore reduce the inflammatory response and improve healing.
Collapse
|
19
|
Strudwick XL, Cowin AJ. Cytoskeletal regulation of dermal regeneration. Cells 2012; 1:1313-27. [PMID: 24710556 PMCID: PMC3901152 DOI: 10.3390/cells1041313] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 11/15/2012] [Accepted: 12/04/2012] [Indexed: 12/21/2022] Open
Abstract
Wound healing results in the repair of injured tissues however fibrosis and scar formation are, more often than not the unfortunate consequence of this process. The ability of lower order vertebrates and invertebrates to regenerate limbs and tissues has been all but lost in mammals; however, there are some instances where glimpses of mammalian regenerative capacity do exist. Here we describe the unlocked potential that exists in mammals that may help us understand the process of regeneration post-injury and highlight the potential role of the actin cytoskeleton in this process. The precise function and regulation of the cytoskeleton is critical to the success of the healing process and its manipulation may therefore facilitate regenerative healing. The gelsolin family of actin remodelling proteins in particular has been shown to have important functions in wound healing and family member Flightless I (Flii) is involved in both regeneration and repair. Understanding the interactions between different cytoskeletal proteins and their dynamic control of processes including cellular adhesion, contraction and motility may assist the development of therapeutics that will stimulate regeneration rather than repair.
Collapse
Affiliation(s)
- Xanthe L Strudwick
- Wound Healing Laboratory, Women's and Children's Health Research Institute, 72 King William Road, North Adelaide, South Australia 5006, Australia.
| | - Allison J Cowin
- Wound Healing Laboratory, Women's and Children's Health Research Institute, 72 King William Road, North Adelaide, South Australia 5006, Australia.
| |
Collapse
|