1
|
Hameedi SG, Saulsbery A, Olutoye OO. The Pathophysiology and Management of Pathologic Scarring-a Contemporary Review. Adv Wound Care (New Rochelle) 2025; 14:48-64. [PMID: 38545753 PMCID: PMC11839539 DOI: 10.1089/wound.2023.0185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Significance: Pathologic scarring occurs secondary to imbalances in the cellular mechanisms of wound healing and affects millions of people annually. This review article aims to provide a concise overview of the pathophysiology and management of pathologic scarring for clinicians and scientists alike. Recent Advances: Contemporary research in the field has identified aberrations in transforming growth factor-β/small mothers against decapentaplegic (TGF-β/SMAD) signaling pathways as key drivers of pathologic scar formation; indeed, this pathway is targeted by many treatment modalities and translational investigations currently underway. Although intralesional injection of corticosteroids has been the gold standard in the treatment of pathologic scarring, studies show greater treatment efficacy with the use of combination injections such as triamcinolone/5-fluorouracil and triamcinolone/botulinum toxin. Adjunctive therapies including ablative fractional carbon dioxide/erbium-doped yttrium aluminum garnet and non-ablative pulsed-dye lasers, microneedling, and carboxytherapy have shown encouraging results in small cohort studies. Translational investigations involving the use of nanogels, RNA interference, and small molecules targeting TGF-β/SMAD pathways are also currently underway and hold promise for the future. Critical Issues: The heterogeneous nature of hypertrophic scars and keloids poses significant challenges in formulating standardized treatment and assessment protocols, thereby limiting the conclusions that can be drawn. Future Directions: Rigorous clinical trials into the individual and synergistic effects of these therapies would be ideal before any definitive conclusions or evidence-based treatment recommendations can be made. Owing to the heterogeneity of the pathology and patient population, well-conducted cohort studies may be the next best option.
Collapse
Affiliation(s)
- Sophia G. Hameedi
- Center for Regenerative Medicine, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Angela Saulsbery
- Center for Regenerative Medicine, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Oluyinka O. Olutoye
- Center for Regenerative Medicine, Nationwide Children’s Hospital, Columbus, Ohio, USA
- Department of Surgery, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
2
|
Machcinska S, Walendzik K, Kopcewicz M, Wisniewska J, Rokka A, Pääkkönen M, Slowinska M, Gawronska-Kozak B. Hypoxia reveals a new function of Foxn1 in the keratinocyte antioxidant defense system. FASEB J 2022; 36:e22436. [PMID: 35792861 DOI: 10.1096/fj.202200249rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 01/12/2023]
Abstract
Skin exposed to environmental threats, including injuries and oxidative stress, develops an efficient but not fully recognized system of repair and antioxidant protection. Here, using mass spectrometry analysis (LC-MS/MS), followed by in vitro and in vivo experiments, we provided evidence that Foxn1 in keratinocytes regulates elements of the electron transport chain and participates in the thioredoxin system (Txn2, Txnrd3, and Srxn1) induction, particularly in a hypoxic environment. We first showed that Foxn1 in keratinocytes upregulates glutathione thioredoxin reductase 3 (Txnrd3) protein expression, and high levels of Txnrd3 mRNA were detected in injured skin of Foxn1+/+ mice. We also showed that Foxn1 strongly downregulated the Ccn2 protein expression, participating in epidermal reconstruction after injury. An in vitro assay revealed that Foxn1 controls keratinocyte migration, stimulating it under normoxia and suppressing it under hypoxia. Keratinocytes overexpressing Foxn1 and exposed to hypoxia displayed a reduced ability to promote angiogenesis by downregulating Vegfa expression. In conclusion, this study showed a new mechanism in which Foxn1, along with hypoxia, participates in the activation of antioxidant defense and controls the functional properties of keratinocytes.
Collapse
Affiliation(s)
- Sylwia Machcinska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Katarzyna Walendzik
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Marta Kopcewicz
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Joanna Wisniewska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Anne Rokka
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Mirva Pääkkönen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Mariola Slowinska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Barbara Gawronska-Kozak
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
3
|
Injectable hydrogels based on silk fibroin peptide grafted hydroxypropyl chitosan and oxidized microcrystalline cellulose for scarless wound healing. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.129062] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
4
|
Parikh UM, Mentz J, Collier I, Davis MJ, Abu-Ghname A, Colchado D, Short WD, King A, Buchanan EP, Balaji S. Strategies to Minimize Surgical Scarring: Translation of Lessons Learned from Bedside to Bench and Back. Adv Wound Care (New Rochelle) 2022; 11:311-329. [PMID: 34416825 DOI: 10.1089/wound.2021.0010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Significance: An understanding of the physiology of wound healing and scarring is necessary to minimize surgical scar formation. By reducing tension across the healing wound, eliminating excess inflammation and infection, and encouraging perfusion to healing areas, surgeons can support healing and minimize scarring. Recent Advances: Preoperatively, newer techniques focused on incision placement to minimize tension, skin sterilization to minimize infection and inflammation, and control of comorbid factors to promote a healing process with minimal scarring are constantly evolving. Intraoperatively, measures like layered closure, undermining, and tissue expansion can be taken to relieve tension across the healing wound. Appropriate suture technique and selection should be considered, and finally, there are new surgical technologies available to reduce tension across the closure. Postoperatively, the healing process can be supported as proliferation and remodeling take place within the wound. A balance of moisture control, tension reduction, and infection prevention can be achieved with dressings, ointments, and silicone. Vitamins and corticosteroids can also affect the scarring process by modulating the cellular factors involved in healing. Critical Issues: Healing with no or minimal scarring is the ultimate goal of wound healing research. Understanding how mechanical tension, inflammation and infection, and perfusion and hypoxia impact profibrotic pathways allows for the development of therapies that can modulate cytokine response and the wound extracellular microenvironment to reduce fibrosis and scarring. Future Directions: New tension-off loading topical treatments, laser, and dermabrasion devices are under development, and small molecule therapeutics have demonstrated scarless wound healing in animal models, providing a promising new direction for future research aimed to minimize surgical scarring.
Collapse
Affiliation(s)
- Umang M. Parikh
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - James Mentz
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Ian Collier
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Matthew J. Davis
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Amjed Abu-Ghname
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Daniel Colchado
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Walker D. Short
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Alice King
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Edward P. Buchanan
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Swathi Balaji
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
5
|
Hirman AR, Du L, Cheng S, Zheng H, Duo L, Zhai Q, Xu J. MiR-133a-3p inhibits scar formation in scalded mice and suppresses the proliferation and migration of scar derived-fibroblasts by targeting connective tissue growth factor. Exp Anim 2021; 70:322-332. [PMID: 33658464 PMCID: PMC8390314 DOI: 10.1538/expanim.20-0159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Excessive scar formation post burn injury can cause great pain to the patients. MiR-133a-3p has been demonstrated to be anti-fibrotic in some fibrosis-related
diseases. However, its possible role in scar formation has not been elucidated yet. In present study, the effect of miR-133a-3p on scar formation was
investigated in a scalded model of mice. Moreover, the function of miR-133a-3p on proliferation and migration of scar-derived fibroblasts (SFs) was studied
in vitro. It was found that miR-133a-3p was dramatically downregulated in scar tissue of scalded mice. Upregulation of miR-133a-3p by
miR-133a-3p agomir obviously inhibited the scar formation in scalded mice. Histological staining showed that upregulation of miR-133a-3p attenuated the
excessive deposition of collagen in scar tissue of scalded mice. In vitro study showed that upregulation of miR-133a-3p effectively suppressed
the proliferation and migration of SFs. Besides, upregulation of miR-133a-3p attenuated the protein levels of α-smooth muscle actin (α-SMA) and collagen I,
indicating that miR-133a-3p could suppress the activation of SFs. The expression of connective tissue growth factor (CTGF), a critical mediator in cell
proliferation, migration and extracellular matrix (ECM) synthesis, was also downregulated by the upregulation of miR-133a-3p. Luciferase reporter assay
validated that CTGF was directly targeted by miR-133a-3p. In addition, overexpression of CTGF abolished the effect of miR-133a-3p on inhibiting the
proliferation, migration and activation of SFs, indicating that miR-133a-3p functioned by targeting CTGF. Therefore, miR-133a-3p might be a promising target for
treating pathological scars.
Collapse
Affiliation(s)
- Abdul Razaq Hirman
- Department of Dermatology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang 110004, P.R. China
| | - Lili Du
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, No. 77, Puhe Road, Shenbei New District, Shenyang 110122, P.R. China
| | - Shaohang Cheng
- Department of Dermatology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang 110004, P.R. China
| | - Heng Zheng
- Department of Dermatology, Central Hospital Affiliated to Shenyang Medical College, No. 7, Nanqi West Road, Tiexi District, Shenyang 110024, P.R. China
| | - Linna Duo
- Department of Dermatology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang 110004, P.R. China
| | - Qianyu Zhai
- Department of Dermatology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang 110004, P.R. China
| | - Jing Xu
- Department of Dermatology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Shenyang 110004, P.R. China
| |
Collapse
|
6
|
Leguit RJ, Raymakers RAP, Hebeda KM, Goldschmeding R. CCN2 (Cellular Communication Network factor 2) in the bone marrow microenvironment, normal and malignant hematopoiesis. J Cell Commun Signal 2021; 15:25-56. [PMID: 33428075 PMCID: PMC7798015 DOI: 10.1007/s12079-020-00602-2] [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: 04/12/2020] [Accepted: 12/20/2020] [Indexed: 02/06/2023] Open
Abstract
CCN2, formerly termed Connective Tissue Growth Factor, is a protein belonging to the Cellular Communication Network (CCN)-family of secreted extracellular matrix-associated proteins. As a matricellular protein it is mainly considered to be active as a modifier of signaling activity of several different signaling pathways and as an orchestrator of their cross-talk. Furthermore, CCN2 and its fragments have been implicated in the regulation of a multitude of biological processes, including cell proliferation, differentiation, adhesion, migration, cell survival, apoptosis and the production of extracellular matrix products, as well as in more complex processes such as embryonic development, angiogenesis, chondrogenesis, osteogenesis, fibrosis, mechanotransduction and inflammation. Its function is complex and context dependent, depending on cell type, state of differentiation and microenvironmental context. CCN2 plays a role in many diseases, especially those associated with fibrosis, but has also been implicated in many different forms of cancer. In the bone marrow (BM), CCN2 is highly expressed in mesenchymal stem/stromal cells (MSCs). CCN2 is important for MSC function, supporting its proliferation, migration and differentiation. In addition, stromal CCN2 supports the maintenance and longtime survival of hematopoietic stem cells, and in the presence of interleukin 7, stimulates the differentiation of pro-B lymphocytes into pre-B lymphocytes. Overexpression of CCN2 is seen in the majority of B-acute lymphoblastic leukemias, especially in certain cytogenetic subgroups associated with poor outcome. In acute myeloid leukemia, CCN2 expression is increased in MSCs, which has been associated with leukemic engraftment in vivo. In this review, the complex function of CCN2 in the BM microenvironment and in normal as well as malignant hematopoiesis is discussed. In addition, an overview is given of data on the remaining CCN family members regarding normal and malignant hematopoiesis, having many similarities and some differences in their function.
Collapse
Affiliation(s)
- Roos J. Leguit
- Department of Pathology, University Medical Center Utrecht, H04-312, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| | - Reinier A. P. Raymakers
- Department of Hematology, UMCU Cancer Center, Heidelberglaan 100 B02.226, 3584 CX Utrecht, The Netherlands
| | - Konnie M. Hebeda
- Department of Pathology, Radboud University Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Centre Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
7
|
Targeting CTGF in Cancer: An Emerging Therapeutic Opportunity. Trends Cancer 2020; 7:511-524. [PMID: 33358571 DOI: 10.1016/j.trecan.2020.12.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022]
Abstract
Despite the dramatic advances in cancer research over the decades, effective therapeutic strategies are still urgently needed. Increasing evidence indicates that connective tissue growth factor (CTGF), a multifunctional signaling modulator, promotes cancer initiation, progression, and metastasis by regulating cell proliferation, migration, invasion, drug resistance, and epithelial-mesenchymal transition (EMT). CTGF is also involved in the tumor microenvironment in most of the nodes, including angiogenesis, inflammation, and cancer-associated fibroblast (CAF) activation. In this review, we comprehensively discuss the expression of CTGF and its regulation, oncogenic role, clinical relevance, targeting strategies, and therapeutic agents. Herein, we propose that CTGF is a promising cancer therapeutic target that could potentially improve the clinical outcomes of cancer patients.
Collapse
|
8
|
Ma D, Chen L, Shi J, Zhao Y, Vasani S, Chen K, Romana‐Souza B, Henkin J, DiPietro LA. Pigment epithelium‐derived factor attenuates angiogenesis and collagen deposition in hypertrophic scars. Wound Repair Regen 2020; 28:684-695. [DOI: 10.1111/wrr.12828] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 04/28/2020] [Accepted: 05/05/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Da Ma
- Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital Guanghua School of Stomatology, SunYat‐sen University Guangzhou Guangdong China
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, College of Dentistry University of Illinois at Chicago Chicago Illinois USA
| | - Junhe Shi
- Center for Wound Healing and Tissue Regeneration, College of Dentistry University of Illinois at Chicago Chicago Illinois USA
| | - Yan Zhao
- Center for Wound Healing and Tissue Regeneration, College of Dentistry University of Illinois at Chicago Chicago Illinois USA
| | - Shruti Vasani
- Center for Wound Healing and Tissue Regeneration, College of Dentistry University of Illinois at Chicago Chicago Illinois USA
| | - Kevin Chen
- Center for Wound Healing and Tissue Regeneration, College of Dentistry University of Illinois at Chicago Chicago Illinois USA
| | - Bruna Romana‐Souza
- Tissue Repair Laboratory State University of Rio de Janeiro Rio de Janeiro Brazil
| | - Jack Henkin
- Center for Developmental Therapeutics and Department of Chemistry Northwestern University Evanston Illinois USA
| | - Luisa A. DiPietro
- Center for Wound Healing and Tissue Regeneration, College of Dentistry University of Illinois at Chicago Chicago Illinois USA
| |
Collapse
|
9
|
Stepien DM, Hwang C, Marini S, Pagani CA, Sorkin M, Visser ND, Huber AK, Edwards NJ, Loder SJ, Vasquez K, Aguilar CA, Kumar R, Mascharak S, Longaker MT, Li J, Levi B. Tuning Macrophage Phenotype to Mitigate Skeletal Muscle Fibrosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:2203-2215. [PMID: 32161098 PMCID: PMC8080967 DOI: 10.4049/jimmunol.1900814] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 01/20/2020] [Indexed: 12/16/2022]
Abstract
Myeloid cells are critical to the development of fibrosis following muscle injury; however, the mechanism of their role in fibrosis formation remains unclear. In this study, we demonstrate that myeloid cell-derived TGF-β1 signaling is increased in a profibrotic ischemia reperfusion and cardiotoxin muscle injury model. We found that myeloid-specific deletion of Tgfb1 abrogates the fibrotic response in this injury model and reduces fibro/adipogenic progenitor cell proliferation while simultaneously enhancing muscle regeneration, which is abrogated by adaptive transfer of normal macrophages. Similarly, a murine TGFBRII-Fc ligand trap administered after injury significantly reduced muscle fibrosis and improved muscle regeneration. This study ultimately demonstrates that infiltrating myeloid cell TGF-β1 is responsible for the development of traumatic muscle fibrosis, and its blockade offers a promising therapeutic target for preventing muscle fibrosis after ischemic injury.
Collapse
Affiliation(s)
- David M Stepien
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Charles Hwang
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Simone Marini
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109
| | - Chase A Pagani
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Michael Sorkin
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Noelle D Visser
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Amanda K Huber
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Nicole J Edwards
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Shawn J Loder
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Kaetlin Vasquez
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Carlos A Aguilar
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109
- Biomedical Engineering Department, Biointerfaces Institute and Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109
| | - Ravi Kumar
- Acceleron Pharmaceuticals, Cambridge MA 02139
| | - Shamik Mascharak
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford University, Stanford, CA 94305; and
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford University, Stanford, CA 94305; and
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford University, Stanford, CA 94305
| | - Jun Li
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109
| | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109;
| |
Collapse
|
10
|
Barbe MF, Hilliard BA, Amin M, Harris MY, Hobson LJ, Cruz GE, Popoff SN. Blocking CTGF/CCN2 reduces established skeletal muscle fibrosis in a rat model of overuse injury. FASEB J 2020; 34:6554-6569. [PMID: 32227398 PMCID: PMC7200299 DOI: 10.1096/fj.202000240rr] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 12/12/2022]
Abstract
Tissue fibrosis is a hallmark of overuse musculoskeletal injuries and contributes to functional declines. We tested whether inhibition of CCN2 (cellular communication network factor 2, previously known as connective tissue growth factor, CTGF) using a specific antibody (termed FG‐3019 or pamrevlumab) reduces established overuse‐induced muscle fibrosis in a clinically relevant rodent model of upper extremity overuse injury. Young adult rats performed a high repetition high force (HRHF) reaching and lever‐pulling task for 18 weeks, after first being shaped for 6 weeks to learn this operant task. Rats were then euthanized (HRHF‐Untreated), or rested and treated for 6 weeks with FG‐3019 (HRHF‐Rest/FG‐3019) or a human IgG as a vehicle control (HRHF‐Rest/IgG). HRHF‐Untreated and HRHF‐Rest/IgG rats had higher muscle levels of several fibrosis‐related proteins (TGFβ1, CCN2, collagen types I and III, and FGF2), and higher muscle numbers of alpha SMA and pERK immunopositive cells, compared to control rats. Each of these fibrogenic changes was restored to control levels by the blocking of CCN2 signaling in HRHF‐Rest/FG‐3019 rats, as were HRHF task‐induced increases in serum CCN2 and pro‐collagen I intact N‐terminal protein. Levels of cleaved CCN3, an antifibrotic protein, were lowered in HRHF‐Untreated and HRHF‐Rest/IgG rats, compared to control rats, yet elevated back to control levels in HRHF‐Rest/FG‐3019 rats. Significant grip strength declines observed in HRHF‐Untreated and HRHF‐Rest/IgG rats, were restored to control levels in HRHF‐Rest/FG‐3019 rats. These results are highly encouraging for use of FG‐3019 for therapeutic treatment of persistent skeletal muscle fibrosis, such as those induced with chronic overuse.
Collapse
Affiliation(s)
- Mary F Barbe
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Brendan A Hilliard
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Mamta Amin
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Michele Y Harris
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Lucas J Hobson
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Geneva E Cruz
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Steven N Popoff
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
11
|
Barbe MF, Hilliard BA, Delany SP, Iannarone VJ, Harris MY, Amin M, Cruz GE, Barreto‐Cruz Y, Tran N, Day EP, Hobson LJ, Assari S, Popoff SN. Blocking CCN2 Reduces Progression of Sensorimotor Declines and Fibrosis in a Rat Model of Chronic Repetitive Overuse. J Orthop Res 2019; 37:2004-2018. [PMID: 31041999 PMCID: PMC6688947 DOI: 10.1002/jor.24337] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 04/12/2019] [Indexed: 02/06/2023]
Abstract
Fibrosis may be a key factor in sensorimotor dysfunction in patients with chronic overuse-induced musculoskeletal disorders. Using a clinically relevant rodent model, in which performance of a high demand handle-pulling task induces tissue fibrosis and sensorimotor declines, we pharmacologically blocked cellular communication network factor 2 (CCN2; connective tissue growth factor) with the goal of reducing the progression of these changes. Young adult, female Sprague-Dawley rats were shaped to learn to pull at high force levels (10 min/day, 5 weeks), before performing a high repetition high force (HRHF) task for 3 weeks (2 h/day, 3 days/week). HRHF rats were untreated, or treated in task weeks 2 and 3 with a monoclonal antibody that blocks CCN2 (FG-3019), or a control immunoglobulin G (IgG). Control rats were untreated or received FG-3019, IgG, or vehicle (saline) injections. Mean task reach rate and grasp force were higher in 3-week HRHF + FG-3019 rats, compared with untreated HRHF rats. Grip strength declined while forepaw mechanical sensitivity increased in untreated HRHF rats, compared with controls; changes improved by FG-3019 treatment. The HRHF task increased collagen in multiple tissues (flexor digitorum muscles, nerves, and forepaw dermis), which was reduced with FG-3019 treatment. FG-3019 treatment also reduced HRHF-induced increases in CCN2 and transforming growth factor β in muscles. In tendons, FG-3019 reduced HRHF-induced increases in CCN2, epitendon thickening, and cell proliferation. Our findings indicate that CCN2 is critical to the progression of chronic overuse-induced multi-tissue fibrosis and functional declines. FG-3019 treatment may be a novel therapeutic strategy for overuse-induced musculoskeletal disorders. © 2019 The Authors. Journal of Orthopaedic Research® published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society. J Orthop Res 37:2004-2018, 2019.
Collapse
Affiliation(s)
- Mary F. Barbe
- Department of Anatomy and Cell Biology, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvania 19140
| | - Brendan A. Hilliard
- Department of Anatomy and Cell Biology, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvania 19140
| | - Sean P. Delany
- Department of Anatomy and Cell Biology, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvania 19140
| | - Victoria J. Iannarone
- Department of Anatomy and Cell Biology, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvania 19140
| | - Michele Y. Harris
- Department of Anatomy and Cell Biology, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvania 19140
| | - Mamta Amin
- Department of Anatomy and Cell Biology, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvania 19140
| | - Geneva E. Cruz
- Department of Anatomy and Cell Biology, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvania 19140
| | - Yeidaliz Barreto‐Cruz
- Department of Anatomy and Cell Biology, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvania 19140
| | - Ngih Tran
- Department of Anatomy and Cell Biology, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvania 19140
| | - Emily P. Day
- Department of Anatomy and Cell Biology, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvania 19140
| | - Lucas J. Hobson
- Department of Anatomy and Cell Biology, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvania 19140
| | - Soroush Assari
- Department of Mechanical Engineering, College of EngineeringTemple UniversityPhiladelphiaPennsylvania 19122
| | - Steven N. Popoff
- Department of Anatomy and Cell Biology, Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvania 19140
| |
Collapse
|
12
|
Coentro JQ, Pugliese E, Hanley G, Raghunath M, Zeugolis DI. Current and upcoming therapies to modulate skin scarring and fibrosis. Adv Drug Deliv Rev 2019; 146:37-59. [PMID: 30172924 DOI: 10.1016/j.addr.2018.08.009] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/08/2018] [Accepted: 08/26/2018] [Indexed: 12/12/2022]
Abstract
Skin is the largest organ of the human body. Being the interface between the body and the outer environment, makes it susceptible to physical injury. To maintain life, nature has endowed skin with a fast healing response that invariably ends in the formation of scar at the wounded dermal area. In many cases, skin remodelling may be impaired, leading to local hypertrophic scars or keloids. One should also consider that the scarring process is part of the wound healing response, which always starts with inflammation. Thus, scarring can also be induced in the dermis, in the absence of an actual wound, during chronic inflammatory processes. Considering the significant portion of the population that is subject to abnormal scarring, this review critically discusses the state-of-the-art and upcoming therapies in skin scarring and fibrosis.
Collapse
Affiliation(s)
- João Q Coentro
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland; Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| | - Eugenia Pugliese
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland; Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| | - Geoffrey Hanley
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland; Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| | - Michael Raghunath
- Center for Cell Biology and Tissue Engineering, Institute for Chemistry and Biotechnology (ICBT), Zurich University of Applied Sciences (ZHAW), Wädenswil, Switzerland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland; Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland.
| |
Collapse
|
13
|
Stechmiller JK, Lyon D, Schultz G, Gibson DJ, Weaver MT, Wilkie D, Ferrell AV, Whitney J, Kim J, Millan SB. Biobehavioral Mechanisms Associated With Nonhealing Wounds and Psychoneurologic Symptoms (Pain, Cognitive Dysfunction, Fatigue, Depression, and Anxiety) in Older Individuals With Chronic Venous Leg Ulcers. Biol Res Nurs 2019; 21:407-419. [PMID: 31142148 DOI: 10.1177/1099800419853881] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The prevalence and incidence of chronic venous leg ulcers (CVLUs) are increasing worldwide, as are the associated financial costs. Although it has long been known that their underlying etiology is venous insufficiency, the molecular aspects of healing versus nonhealing, as well as the psychoneurologic symptoms (PNS; pain, cognitive dysfunction, fatigue, depression, and anxiety) associated with CVLUs remain understudied. In this biobehaviorally focused review, we aim to elucidate the complex mechanisms that link the biological and molecular aspects of CLVUs with their PNS. Innovations in "omics" research have increased our understanding of important wound microenvironmental factors (e.g., inflammation, microbial pathogenic biofilm, epigenetic processes) that may adversely alter the wound bed's molecular milieu so that microbes evade immune detection. Although these molecular factors are not singularly responsible for wound healing, they are major components of wound development, nonhealing, and PNS that, until now, have not been amenable to systematic study, especially over time. Further, this review explores our current understanding of the molecular mechanisms by which the immune activation that contributes to the development and persistence of CVLUs also leads to the development, persistence, and severity of wound-related PNS. We also make recommendations for future research that will expand the field of biobehavioral wound science. Biobehavioral research that focuses on the interrelated mechanisms of PNS will lead to symptom-management interventions that improve quality of life for the population burdened by CVLUs.
Collapse
Affiliation(s)
- Joyce K Stechmiller
- 1 Department of Biobehavioral Nursing Science, College of Nursing, University of Florida, Gainesville, FL, USA
| | - Debra Lyon
- 2 College of Nursing, University of Florida, Gainesville, FL, USA
| | - Gregory Schultz
- 3 Department of Obstetrics and Gynecology, Institute for Wound Research, University of Florida, Gainesville, FL, USA
| | - Daniel J Gibson
- 3 Department of Obstetrics and Gynecology, Institute for Wound Research, University of Florida, Gainesville, FL, USA
| | - Michael T Weaver
- 2 College of Nursing, University of Florida, Gainesville, FL, USA
| | - Diana Wilkie
- 4 Center for Palliative Care Research and Education, University of Florida, Gainesville, FL, USA
| | | | - Joanne Whitney
- 5 School of Nursing, Harborview Medical Center, University of Washington, Seattle, WA, USA
| | - Junglyun Kim
- 2 College of Nursing, University of Florida, Gainesville, FL, USA
| | - Susan B Millan
- 6 UF Health Wound Care and Hyperbaric Center, Gainesville, FL, USA
| |
Collapse
|
14
|
Gonçalves LR, de Oliveira da Silva B, Scarinci LD, Ramos LF, Moraes KC. MicroRNA-1254 contributes to the controlling of pro-fibrogenic environment in LX-2 cells by modulating SMAD3 and wound repair: new insights in hepatic fibrosis. Cell Biol Int 2019; 43:333-343. [DOI: 10.1002/cbin.11075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 11/15/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Letícia Rocha Gonçalves
- Laboratório de Biologia Molecular, Departamento de Biologia; Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho” − Campus Rio Claro; Rio Claro SP 13506-900 Brazil
| | | | - Letícia Destefani Scarinci
- Laboratório de Biologia Molecular, Departamento de Biologia; Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho” − Campus Rio Claro; Rio Claro SP 13506-900 Brazil
| | - Letícia Ferreira Ramos
- Laboratório de Biologia Molecular, Departamento de Biologia; Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho” − Campus Rio Claro; Rio Claro SP 13506-900 Brazil
| | - Karen C.M. Moraes
- Laboratório de Biologia Molecular, Departamento de Biologia; Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho” − Campus Rio Claro; Rio Claro SP 13506-900 Brazil
| |
Collapse
|
15
|
Anti-CTGF Oligonucleotide Reduces Severity of Postsurgical Hypertrophic Scars in a Randomized, Double-Blind, Within-Subject, Placebo-Controlled Study. Plast Reconstr Surg 2019; 142:192e-201e. [PMID: 30045185 DOI: 10.1097/prs.0000000000004590] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Connective tissue growth factor (CTGF) levels are up-regulated in wounded skin and are thought to play a major role in scar formation. An antisense oligonucleotide targeting CTGF was evaluated in adult patients undergoing hypertrophic scar revision surgery, to determine effects on reducing the severity of subsequent scars. METHODS In a randomized, double-blind, within-subject, placebo-controlled study, 23 female subjects (aged 28 to 55 years) with bilateral, symmetric, hypertrophic surgical scars of the breast underwent scar revision surgery. The resulting breast incisions were randomized to receive EXC 001 (5 mg/cm) or placebo injected intradermally at postsurgery weeks 2, 5, 8, and 11. Scar severity assessments were performed at weeks 12 and 24 by an expert panel using blinded photographs, and by physicians and subjects using a scar scoring scale, the Patient and Observer Scar Assessment Scale. An assumption of the design is that within-subject variance would be small and that whatever within-subject variance there was would be controlled through the randomization process. RESULTS EXC 001 significantly reduced scar severity at both 12 and 24 weeks after scar revision surgery in all three measures (expert panel and physician Patient and Observer Scar Assessment Scale, p < 0.001; Patient and Observer Scar Assessment Scale, p < 0.003). CONCLUSIONS This study provided positive preliminary data that intradermal injection of EXC 001 produced a significant reduction in severity of postsurgical skin scars, as measured by physicians, subjects, and an expert panel. This study provided evidence that suppression of CTGF could be a viable strategy for hypertrophic scar reduction therapy and that further study of the antisense oligonucleotide EXC 001 was indicated. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, II.
Collapse
|
16
|
Therapeutic potential of endogenous stem cells and cellular factors for scar-free skin regeneration. Drug Discov Today 2019; 24:69-84. [DOI: 10.1016/j.drudis.2018.10.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 08/28/2018] [Accepted: 10/25/2018] [Indexed: 12/20/2022]
|
17
|
Hairpin-structured probe conjugated nano-graphene oxide for the cellular detection of connective tissue growth factor mRNA. Anal Chim Acta 2018; 1038:140-147. [DOI: 10.1016/j.aca.2018.07.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 07/05/2018] [Accepted: 07/08/2018] [Indexed: 11/22/2022]
|
18
|
Cho Lee AR, Woo I. Local Silencing of Connective Tissue Growth Factor by siRNA/Peptide Improves Dermal Collagen Arrangements. Tissue Eng Regen Med 2018; 15:711-719. [PMID: 30603590 DOI: 10.1007/s13770-018-0166-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/17/2018] [Accepted: 10/19/2018] [Indexed: 12/24/2022] Open
Abstract
Background Collagen organization within tissues has a critical role in wound regeneration. Collagen fibril diameter, arrangements and maturity between connective tissue growth factor (CTGF) small interfering RNA (siRNA) and mismatch scrambled siRNA-treated wound were compared to evaluate the efficacy of CTGF siRNA as a future implement for scar preventive medicine. Methods Nanocomplexes of CTGF small interfering RNA (CTGF siRNA) with cell penetrating peptides (KALA and MPG∆NLS) were formulated and their effects on CTGF downregulation, collagen fibril diameter and arrangement were investigated. Various ratios of CTGF siRNA and peptide complexes were prepared and down-regulation were evaluated by immunoblot analysis. Control and CTGF siRNA modified cells-populated collagen lattices were prepared and rates of contraction measured. Collagen organization in rabbit ear 8 mm biopsy punch wound at 1 day to 8 wks post injury time were investigated by transmission electron microscopy and histology was investigated with Olympus System and TS-Auto software. Conclusion CTGF expression was down-regulated to 40% of control by CTGF siRNA/KALA (1:24) complexes (p < 0.01) and collagen lattice contraction was inhibited. However, down-regulated of CTGF by CTGF siRNA/MPG∆NLS complexes was not statistically significant. CTGF KALA-treated wound appeared with well formed-basket weave pattern of collagen fibrils with mean diameter of 128 ± 22 nm (n = 821). Mismatch siRNA/KALA-treated wound showed a high frequency of parallel small diameter fibrils (mean 90 ± 20 nm, n = 563). Conclusion Controlling over-expression of CTGF by peptide-mediated siRNA delivery could improve the collagen orientation and tissue remodeling in full thickness rabbit ear wound.
Collapse
Affiliation(s)
- Ae-Ri Cho Lee
- College of Pharmacy, Duksung Women's University, 33 Samyang-ro 144-gil, Dobong-gu, Seoul, 01369 Republic of Korea
| | - Inhae Woo
- College of Pharmacy, Duksung Women's University, 33 Samyang-ro 144-gil, Dobong-gu, Seoul, 01369 Republic of Korea
| |
Collapse
|
19
|
A Placebo-controlled Study of PF-06473871 (Anti-Connective Tissue Growth Factor Antisense Oligonucleotide) in Reducing Hypertrophic Skin Scarring. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2018; 6:e1861. [PMID: 30349773 PMCID: PMC6191237 DOI: 10.1097/gox.0000000000001861] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/14/2018] [Indexed: 12/03/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Connective tissue growth factor (CTGF) is a matricellular protein that plays a key role in wound healing and scar formation. Inhibition of CTGF by a specific antisense oligonucleotide significantly reduced scarring and fibrosis in animal models. This study examined whether an antisense oligonucleotide that inhibits human CTGF expression could reduce the severity of hypertrophic scar formation in patients following surgical revision of preexisting breast scars. Methods: This study was a 24-week multicenter, randomized, double-blind, within-subject, placebo-controlled phase 2b study evaluating the efficacy and safety of PF-06473871 in 2 regimens of either 3 or 4 intradermal injections (postsurgery weeks 2, 5, 8, and 11) of 5 mg/cm adjacent to the new surgical incision. One hundred subjects with bilateral hypertrophic scars resulting from prior breast surgery were randomized. Efficacy was determined by the Patient and Observer Scar Assessment Scale (POSAS). Results: The Physician/Observer POSAS overall opinion score at (week 24) for the 4-injection regimen demonstrated a statistically significant (P = 0.022) treatment difference from placebo of 0.68, and the treatment difference for the 3-injection regimen was nonsignificant (P = 0.4). Physician evaluation of scar severity at (week 24) with the photo-guide in the 4-injection regimen had a significant reduction (point estimate of treatment difference of 0.43 favoring PF-06473871). The surgical effect was approximately 2.0 at week 24 and was nearly 3 times greater than the treatment effect. Patient evaluations using the POSAS and photo-guide were not significantly improved with either dose regimen. PF-06473871 was generally well tolerated systemically and locally. Conclusion: The 4-dose regimen of PF-06473871 provided statistically significant improvement, inhibiting severity of hypertrophic scar formation based on physician assessment. However, the effect of revision surgery alone is significant and may dominate the treatment effect of PF-06473871.
Collapse
|
20
|
Siracusa R, Impellizzeri D, Cordaro M, Gugliandolo E, Peritore AF, Di Paola R, Cuzzocrea S. Topical Application of Adelmidrol + Trans-Traumatic Acid Enhances Skin Wound Healing in a Streptozotocin-Induced Diabetic Mouse Model. Front Pharmacol 2018; 9:871. [PMID: 30190675 PMCID: PMC6115498 DOI: 10.3389/fphar.2018.00871] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/18/2018] [Indexed: 12/14/2022] Open
Abstract
Impaired wound healing is considered to be one of the severe complications associated with diabetes. Adelmidrol and trans-traumatic acid are commonly called Nevamast®. This gel consists precisely of 2% adelmidrol and 1% trans-traumatic acid. Thanks to its components, it is capable of favoring the natural process of skin re-epithelialization. This study tests the theory that topical usage of adelmidrol + trans-traumatic acid has important effects on the healing and closure of diabetic wounds in a streptozotocin (STZ)-induced diabetic mouse model. Diabetes was induced by intraperitoneal injection of STZ (60 mg/kg) in 0.01 M citrate buffer (pH 4.5) administrated for 5 consecutive days. After diabetes induction, two longitudinal incisions were made on the dorsum of the mice. The animals were killed between 6 and 12 days from wound induction. We found that diabetic mice compared to control mice presented: a retarded wound closure, characterized by an important reduction in the levels of transforming growth factor-β, plus an important increase of vascular endothelial growth factor and endothelial-type nitric oxide synthase expression, together with a reduction of adhesion molecules such as intercellular adhesion molecule-1 and P-selectin and a prolonged elevation of the levels of matrix metalloproteinase-9 and matrix metalloproteinase-2 in wound tissues. This study demonstrates that topical application of adelmidrol + trans-traumatic acid has important effects on the healing and closure of diabetic wounds in an STZ-induced diabetic mouse model.
Collapse
Affiliation(s)
- Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Marika Cordaro
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Enrico Gugliandolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Alessio F Peritore
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy.,Department of Pharmacological and Physiological Sciences, Saint Louis University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
21
|
Qian W, Li N, Cao Q, Fan J. Thrombospondin-4 critically controls transforming growth factor β1 induced hypertrophic scar formation. J Cell Physiol 2018; 234:731-739. [PMID: 30132849 DOI: 10.1002/jcp.26877] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/18/2018] [Indexed: 12/11/2022]
Abstract
Transforming growth factor β (TGF-β) is a growth factor presenting important functions during tissue remodeling and hypertrophic scar (HS) formation. However, the underlying molecular mechanisms are largely unknown. In this study, we identified thrombospondin-4 (TSP-4) as a TGF-β1 target that essentially mediates TGF-β1-induced scar formation both in vitro and in vivo. The expression of TSP-4 was compared on both mRNA and protein levels between hypertrophic scar fibroblasts (HSFs) and normal skin fibroblast (NFs) in response to TGF-β1 treatment. Two signaling molecules, Smad3 and p38, were assessed for their importance in regulating TGF-β1-mediated TSP-4 expression. The significance of TSP-4 in controlling TGF-β1-induced proliferation, invasion, migration, and fibrosis in HSFs was analyzed by knocking down endogenous TSP-4 using small hairpin RNA (shRNA) (TSP-4 shRNA). Finally, a skin HS model was established in rats and the scar formation was compared between rats treated with vehicle (saline), TGF-β1, and TGF-β1 + TSP-4 shRNA. The TSP-4 level was significantly higher in HSFs than in NFs and TGF-β1 more potently boosted TSP-4 expression in the former than in the latter. Both Smad3 and p38 essentially mediated TGF-β1-induced TSP-4 expression. TSP-4 shRNA significantly suppressed TGF-β1-stimulated proliferation, invasion, migration, or fibrosis of HSFs in vitro and drastically improved wound healing in vivo. TGF-β1, by activating both Smad3 and p38, induces TSP-4, which in turn not only presents a positive feedback regulation on the activation of Smad3 and p38, but also essentially mediates TGF-β1-induced HS formation. Targeting TSP-4 thus may benefit HS treatment.
Collapse
Affiliation(s)
- Wei Qian
- Department of Plastic and Reconstructive Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ning Li
- Department of Plastic and Reconstructive Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Qian Cao
- Department of Plastic and Reconstructive Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jufeng Fan
- Department of Plastic and Reconstructive Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
March JT, Golshirazi G, Cernisova V, Carr H, Leong Y, Lu-Nguyen N, Popplewell LJ. Targeting TGFβ Signaling to Address Fibrosis Using Antisense Oligonucleotides. Biomedicines 2018; 6:biomedicines6030074. [PMID: 29941814 PMCID: PMC6164894 DOI: 10.3390/biomedicines6030074] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 12/29/2022] Open
Abstract
Fibrosis results from the excessive accumulation of extracellular matrix in chronically injured tissue. The fibrotic process is governed by crosstalk between many signaling pathways. The search for an effective treatment is further complicated by the fact that there is a degree of tissue-specificity in the pathways involved, although the process is not completely understood for all tissues. A plethora of drugs have shown promise in pre-clinical models, which is not always borne out translationally in clinical trial. With the recent approvals of two antisense oligonucleotides for the treatment of the genetic diseases Duchenne muscular dystrophy and spinal muscular atrophy, we explore here the potential of antisense oligonucleotides to knockdown the expression of pro-fibrotic proteins. We give an overview of the generalized fibrotic process, concentrating on key players and highlight where antisense oligonucleotides have been used effectively in cellular and animal models of different fibrotic conditions. Consideration is given to the advantages antisense oligonucleotides would have as an anti-fibrotic therapy alongside factors that would need to be addressed to improve efficacy. A prospective outlook for the development of antisense oligonucleotides to target fibrosis is outlined.
Collapse
Affiliation(s)
- James T March
- Centre for Gene and Cell Therapy, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| | - Golnoush Golshirazi
- Centre for Gene and Cell Therapy, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| | - Viktorija Cernisova
- Centre for Gene and Cell Therapy, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| | - Heidi Carr
- Centre for Gene and Cell Therapy, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| | - Yee Leong
- Centre for Gene and Cell Therapy, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| | - Ngoc Lu-Nguyen
- Centre for Gene and Cell Therapy, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| | - Linda J Popplewell
- Centre for Gene and Cell Therapy, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| |
Collapse
|
23
|
Vilela P, Heuer-Jungemann A, El-Sagheer A, Brown T, Muskens OL, Smyth NR, Kanaras AG. Sensing of Vimentin mRNA in 2D and 3D Models of Wounded Skin Using DNA-Coated Gold Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1703489. [PMID: 29464860 DOI: 10.1002/smll.201703489] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/11/2018] [Indexed: 06/08/2023]
Abstract
Wound healing is a highly complex biological process, which is accompanied by changes in cell phenotype, variations in protein expression, and the production of active biomolecules. Currently, the detection of proteins in cells is done by immunostaining where the proteins in fixed cells are detected by labeled antibodies. However, immunostaining cannot provide information about dynamic processes in living cells, within the whole tissue. Here, an easy method is presented to detect the transition of epithelial to mesenchymal cells during wound healing. The method employs DNA-coated gold nanoparticle fluorescent nanoprobes to sense the production of Vimentin mRNA expressed in mesenchymal cells. Fluorescence microscopy is used to achieve temporal detection of Vimentin mRNA in wounds. 3D light-sheet microscopy is utilized to observe the dynamic expression of Vimentin mRNA spatially around the wounded site in skin tissue. The use of DNA-gold nanoprobes to detect mRNA expression during wound healing opens up new possibilities for the study of real-time mechanisms in complex biological processes.
Collapse
Affiliation(s)
- Patrick Vilela
- Physics and Astronomy, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton, SO17 1BJ, UK
| | - Amelie Heuer-Jungemann
- Physics and Astronomy, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton, SO17 1BJ, UK
| | - Afaf El-Sagheer
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, UK
- Chemistry Branch, Department of Science and Mathematics, Faculty of Petroleum and Mining Engineering, Suez University, Suez, 43721, Egypt
| | - Tom Brown
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, UK
| | - Otto L Muskens
- Physics and Astronomy, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton, SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Neil R Smyth
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Antonios G Kanaras
- Physics and Astronomy, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton, SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| |
Collapse
|
24
|
Gauer S, Holzmann Y, Kränzlin B, Hoffmann SC, Gretz N, Hauser IA, Goppelt-Struebe M, Geiger H, Obermüller N. CTGF Is Expressed During Cystic Remodeling in the PKD/Mhm (cy/+) Rat Model for Autosomal-Dominant Polycystic Kidney Disease (ADPKD). J Histochem Cytochem 2017; 65:743-755. [PMID: 29058957 DOI: 10.1369/0022155417735513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Connective tissue growth factor (CTGF, also named CCN2) plays an important role in the development of tubulointerstitial fibrosis, which most critically determines the progression to end-stage renal failure in autosomal-dominant polycystic kidney disease (ADPKD), the most common genetically caused renal disease. We determined CTGF expression in a well-characterized animal model of human ADPKD, the PKD/Mhm (cy/+) rat. Kidneys of 12 weeks old (cy/+) as well as (+/+) non-affected rats were analyzed for CTGF RNA and protein expression by RT-PCR, Northern and Western blot analyses, in situ hybridization, and IHC. Besides the established expression of CTGF in glomerular cells in kidneys of wild-type (+/+) animals, in (cy/+) rats, CTGF mRNA and protein were robustly expressed in interstitial, stellate-shaped cells, located in a scattered pattern underlying the cystic epithelium and in focal areas of advanced tubulointerstitial remodeling. Renal CTGF mRNA and protein expression levels were significantly higher in (cy/+) rats compared with their (+/+) littermates. Detection of CTGF expression in cells adjacent to cystic epithelium and in areas of marked fibrosis suggests a role in the local response to cyst development and indicates that CTGF may be a relevant factor contributing to tubulointerstitial fibrosis in polycystic kidney disease.
Collapse
Affiliation(s)
- Stefan Gauer
- Department of Nephrology, Medical Clinic III, University Hospital Frankfurt, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Yvonne Holzmann
- Department of Nephrology, Medical Clinic III, University Hospital Frankfurt, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Bettina Kränzlin
- Medical Research Center, University of Heidelberg, Mannheim, Germany
| | - Sigrid C Hoffmann
- Medical Research Center, University of Heidelberg, Mannheim, Germany
| | - Norbert Gretz
- Medical Research Center, University of Heidelberg, Mannheim, Germany
| | - Ingeborg A Hauser
- Department of Nephrology, Medical Clinic III, University Hospital Frankfurt, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Margarete Goppelt-Struebe
- Department of Nephrology and Hypertension, Medical Clinic 4, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Helmut Geiger
- Department of Nephrology, Medical Clinic III, University Hospital Frankfurt, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Nicholas Obermüller
- Department of Nephrology, Medical Clinic III, University Hospital Frankfurt, Goethe-University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
25
|
Local Application of Statins Significantly Reduced Hypertrophic Scarring in a Rabbit Ear Model. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2017; 5:e1294. [PMID: 28740761 PMCID: PMC5505822 DOI: 10.1097/gox.0000000000001294] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 02/15/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND We previously showed that intradermal injection of statins is a successful treatment for hypertrophic scarring. Topical application has many advantages over intradermal injection. In this study, we demonstrate the efficacy of topical statin treatment in reducing scar in our validated rabbit ear scar model. METHODS Twenty New Zealand White rabbits were divided into 2 study groups, with 6 rabbits receiving 10 μm pravastatin intradermally at postoperative days 15, 18, and 21, and 14 rabbits receiving 0.4%, 2%, and 10% simvastatin topical application at postoperative days 14-25. Four or 6 full-thickness circular dermal punches 7 mm in diameter were made on the ventral surface of the ear down to but not including the perichondrium. Specimens were collected at 28 days to evaluate the effects of statins on hypertrophic scarring. RESULTS Treatment with pravastatin intradermal administration significantly reduced scarring in terms of scar elevation index. Topical treatment with both medium- and high-dose simvastatin also significantly reduced scarring. High-dose simvastatin topical treatment showed a major effect in scar reduction but induced side effects of scaling, erythema, and epidermal hyperplasia, which were improved with coapplication of cholesterol. There is a dose response in scar reduction with low-, medium- and high-dose simvastatin topical treatment. High-dose simvastatin treatment significantly reduced the messenger ribonucleic acid (mRNA) expression of connective tissue growth factor, consistent with our previously published work on intradermally injected statins. More directly, high-dose simvastatin treatment also significantly reduced the mRNA expression of collagen 1A1. CONCLUSIONS Topical simvastatin significantly reduces scar formation. The mechanism of efficacy for statin treatment through interference with connective tissue growth factor mRNA expression was confirmed.
Collapse
|
26
|
Cho KH, Singh B, Maharjan S, Jang Y, Choi YJ, Cho CS. Local Delivery of CTGF siRNA with Poly(sorbitol-co-PEI) Reduces Scar Contraction in Cutaneous Wound Healing. Tissue Eng Regen Med 2017; 14:211-220. [PMID: 30603478 PMCID: PMC6171600 DOI: 10.1007/s13770-017-0059-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 04/18/2017] [Accepted: 04/24/2017] [Indexed: 10/19/2022] Open
Abstract
Healing process in scarring inevitably produces a considerable amount of non-organized dense collagen-rich matrix called scar thus impairing the native structure of skin. Connective tissue growth factor (CTGF) overexpression within healing tissues is known to play an imperative role in collagen production stimulated by transforming growth factor-beta in cutaneous wound healing. Undoubtedly, the knockdown of CTGF expression through siRNA-mediated gene silencing could simply impede the scarring process. However, the less stability and low transfection of siRNAs themselves urge a safe carrier to protect and transfect them into cells at a high rate avoiding toxicities. Here, we developed a degradable poly(sorbitol-co-PEI) (PSPEI), prepared by polymerization of sorbitol diacrylate with low molecular weight polyethylenimine, which has high transfection efficiency but low cytotoxicity, and utilized it in siCTGF delivery to silence the expression of CTGF in an animal model of cutaneous wound healing. Unlike contracted scar in normal healing, there was no or less contraction in the healed skin of mice treated with siCTGF using PSPEI. Histologically, the healed tissues also had distinct papillary structures and dense irregular connective tissues that were lacking in the control scar tissues. This study exemplifies a successful treatment of cutaneous wound healing using a polymer system coupled with RNA interference. Hence, the approach holds a great promise for developing new treatments with novel targets in regenerative medicines.
Collapse
Affiliation(s)
- Ki-Hyun Cho
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826 Korea
| | - Bijay Singh
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826 Korea
- Research Institute for Bioscience and Biotechnology, Kathmandu, 44600 Nepal
| | - Sushila Maharjan
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826 Korea
- Research Institute for Bioscience and Biotechnology, Kathmandu, 44600 Nepal
| | - Yoonjeong Jang
- Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul, 08826 Korea
| | - Yun-Jaie Choi
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826 Korea
| | - Chong-Su Cho
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826 Korea
| |
Collapse
|
27
|
Gibson DJ, Tuli SS, Schultz GS. Dual-Phase Iontophoresis for the Delivery of Antisense Oligonucleotides. Nucleic Acid Ther 2017; 27:238-250. [PMID: 28375679 DOI: 10.1089/nat.2016.0654] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In support of ongoing research in the study of corneal and skin wound healing, we sought to improve on previously published results by using iontophoresis to deliver RNA interference-based oligonucleotides. By using a electromechanics-based approach, we were able to devise a two-phase solution that separated the buffering solution from the antisense oligonucleotide (ASO) solution. The separation was obtained by making the drug solution a higher density than the buffer, leading it to sink directly onto the tissue surface. This change immediately decreased the distance that the ASO would have to travel before delivery. The changes enabled delivery into ex vivo skin and corneas in 10 or fewer minutes and into in vivo corneas in 5 min. In vivo studies demonstrated short-term bioavailability of at least 24 h, a lack of chemical or thermal injury, a lack of interference in the healing of a corneal injury, and an antisense effect till at least day 7, but not day 14. The only side-effect observed was postdelivery edema that was not present when the vehicle alone was iontophoresed. This suggests that electro-osmotic flow from the delivery chamber was not the mechanism, but that the delivered solute likely increased the tissue's osmolarity. These results support the continued development and utilization of this ASO delivery approach in research-grade oligonucleotides to probe molecular biological pathways and in support of testing therapeutic ASOs in the skin and cornea.
Collapse
Affiliation(s)
- Daniel J Gibson
- 1 Institute for Wound Research, University of Florida , Gainesville, Florida
| | - Sonal S Tuli
- 2 Department of Ophthalmology, University of Florida , Gainesville, Florida
| | - Gregory S Schultz
- 1 Institute for Wound Research, University of Florida , Gainesville, Florida
| |
Collapse
|
28
|
TMF and glycitin act synergistically on keratinocytes and fibroblasts to promote wound healing and anti-scarring activity. Exp Mol Med 2017; 49:e302. [PMID: 28303029 PMCID: PMC5382558 DOI: 10.1038/emm.2016.167] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 09/27/2016] [Accepted: 10/31/2016] [Indexed: 01/04/2023] Open
Abstract
Keratinocyte-fibroblast interactions are critical for skin repair after injury. During the proliferative phase of wound healing, proliferation, migration and differentiation of these cells are the major mechanisms leading to tissue remodeling. We have previously reported that glycitin, a major soy isoflavone, stimulates dermal fibroblast proliferation; and the phytochemical, 4′,6,7-trimethoxyisoflavone (TMF), induces migration of HaCaT keratinocyte cells. We therefore investigated whether these compounds display synergistic effects on skin cells during wound healing in vitro and in vivo. Co-treatment with TMF and glycitin synergistically promotes the proliferation and migration of both keratinocytes and dermal fibroblasts, with a 1:1 ratio of these compounds showing the greatest efficacy in our co-culture system. This keratinocyte-fibroblast interaction occurred via the secretion of TGF-β, and the induction of differentiation and proliferation was confirmed in both indirect and direct co-culture assays. In an excisional and burn wound animal model, mice treated with a 1:1 ratio of TMF and glycitin showed faster wound closure, regeneration and scar reduction than even the positive control drug. These data indicate that two isoflavones, TMF and glycitin, act synergistically to promote wound healing and anti-scarring and could potentially be developed together as a bioactive therapeutic for wound treatment.
Collapse
|
29
|
Shen C, Jiang L, Shao H, You C, Zhang G, Ding S, Bian T, Han C, Meng Q. Targeted killing of myofibroblasts by biosurfactant di-rhamnolipid suggests a therapy against scar formation. Sci Rep 2016; 6:37553. [PMID: 27901027 PMCID: PMC5128858 DOI: 10.1038/srep37553] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 11/01/2016] [Indexed: 01/06/2023] Open
Abstract
Pathological myofibroblasts are often involved in skin scarring via generating contractile force and over-expressing collagen fibers, but no compound has been found to inhibit the myofibroblasts without showing severe toxicity to surrounding physiological cells. Here we report that di-rhamnolipid, a biosurfactant secreted by Pseudomonas aeruginosa, showed potent effects on scar therapy via a unique mechanism of targeted killing the myofibroblasts. In cell culture, the fibroblasts-derived myofibroblasts were more sensitive to di-rhamnolipid toxicity than fibroblasts at a concentration-dependent manner, and could be completely inhibited of their specific functions including α-SMA expression and collagen secretion/contraction. The anti-fibrotic function of di-rhamnolipid was further verified in rabbit ear hypertrophic scar models by presenting the significant reduction of scar elevation index, type I collagen fibers and α-SMA expression. In this regard, di-rhamnolipid treatment could be suggested as a therapy against skin scarring.
Collapse
Affiliation(s)
- Chong Shen
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, PR China
| | - Lifang Jiang
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, PR China
| | - Huawei Shao
- Department of Burns &Wound Care Centre, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, PR China
| | - Chuangang You
- Department of Burns &Wound Care Centre, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, PR China
| | - Guoliang Zhang
- Ocean College, Zhejiang University of Technology, Hangzhou, PR China
| | - Sitong Ding
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, PR China
| | - Tingwei Bian
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, PR China
| | - Chunmao Han
- Department of Burns &Wound Care Centre, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, PR China
| | - Qin Meng
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, PR China
| |
Collapse
|
30
|
S100A12 Induced in the Epidermis by Reduced Hydration Activates Dermal Fibroblasts and Causes Dermal Fibrosis. J Invest Dermatol 2016; 137:650-659. [PMID: 27840235 DOI: 10.1016/j.jid.2016.10.040] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 10/05/2016] [Accepted: 10/26/2016] [Indexed: 12/16/2022]
Abstract
Disruption of the barrier function of skin increases transepidermal water loss and up-regulates inflammatory pathways in the epidermis. Consequently, sustained expression of proinflammatory cytokines from the epidermis is associated with dermal scarring. We found increased expression of S100A12 in the epidermis of human hypertrophic and keloid scar. Exposing a stratified keratinocyte culture to a reduced-hydration environment increased the expression and secretion of S100A12 by nearly 70%, which in turn activated dermal fibroblasts in vitro. Direct treatment of fibroblasts with conditioned medium collected from stratified keratinocyte culture under reduced-hydration conditions activated fibroblasts, shown by up-regulation of α-smooth muscle actin, pro-collagen 1, and F-actin expression. However, this fibroblast activation was not found when S100A12 was knocked down by RNA interference in keratinocytes. Pharmacological blockade of S100A12 receptors, RAGE, or TLR4 inhibited S100A12-induced fibroblast activation. Local delivery of S100A12 resulted in a marked hypertrophic scar formation in a validated rabbit hypertrophic scar model compared with saline control. Our findings indicate that S100A12 functions as a proinflammatory cytokine and suggest that S100A12 is a potential therapeutic target for dermal scarring.
Collapse
|
31
|
Growth factor pathways in hypertrophic scars: Molecular pathogenesis and therapeutic implications. Biomed Pharmacother 2016; 84:42-50. [PMID: 27636511 DOI: 10.1016/j.biopha.2016.09.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 08/26/2016] [Accepted: 09/05/2016] [Indexed: 01/07/2023] Open
Abstract
Hypertrophic scars represent the most common complication of skin injury and are caused by excessive cutaneous wound healing characterized by hypervascularity and pathological deposition of extracellular matrix (ECM) components. To date, the optimal and specific treatment methods for hypertrophic scars have not been available in the clinic. Current paradigm has established fibroblasts and myofibroblasts as pivotal effector cells in the pathophysiology of wound healing. Their biological properties including origin, proliferation, migration, contraction and ECM regulation have profound impacts on the progression and regression of hypertrophic scars. These complex processes are executed and modulated by a signaling network involving a number of growth factors and cytokines. Of particular importance is transforming growth factor-β, platelet-derived growth factor, connective tissue growth factor, epidermal growth factor, and vascular endothelial growth factor. This review article briefly describes the biological functions of fibroblasts and myofibroblasts during hypertrophic scars, and thereafter examines the up-to-date molecular knowledge on the roles of key growth factor pathways in the pathophysiology of hypertrophic scars. Importantly, the therapeutic implications and future challenges of these molecular discoveries are critically discussed in the hope of advancing therapeutic approaches to limit pathological scar formation.
Collapse
|
32
|
Development of Cell-Penetrating Asymmetric Interfering RNA Targeting Connective Tissue Growth Factor. J Invest Dermatol 2016; 136:2305-2313. [PMID: 27427487 DOI: 10.1016/j.jid.2016.06.626] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 06/10/2016] [Accepted: 06/16/2016] [Indexed: 01/12/2023]
Abstract
Connective tissue growth factor (CTGF) is a multifunctional matricellular protein, playing a role as a central mediator in tissue remodeling and fibrosis. A number of reports have shown the pivotal roles of CTGF in the progression of fibrosis, suggesting CTGF as a promising therapeutic target for the treatment of fibrotic disorders including hypertrophic scars and keloids. In this study, we present the development of an interfering RNA molecule that efficiently inhibits the expression of CTGF via RNA interference mechanism both in vitro and in vivo. Chemical modifications were introduced to the asymmetric interfering RNA (asiRNA) backbone structure. The resulting RNA molecule, termed cell-penetrating asiRNA (cp-asiRNA), entered into cells and triggered RNA interference-mediated gene silencing without delivery vehicles. The gene-silencing activity of cp-asiRNA targeting CTGF (cp-asiCTGF) was examined both in vitro and in vivo. Furthermore, the administration of cp-asiCTGF in the rat skin excision wound model efficiently reduced the induction of CTGF and collagens during the wound-healing process. These results suggest that the cp-asiCTGF molecule could be developed into antifibrotic therapeutics such as antiscar drugs.
Collapse
|
33
|
Mechanoregulation of Wound Healing and Skin Homeostasis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3943481. [PMID: 27413744 PMCID: PMC4931093 DOI: 10.1155/2016/3943481] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/10/2016] [Indexed: 02/06/2023]
Abstract
Basic and clinical studies on mechanobiology of cells and tissues point to the importance of mechanical forces in the process of skin regeneration and wound healing. These studies result in the development of new therapies that use mechanical force which supports effective healing. A better understanding of mechanobiology will make it possible to develop biomaterials with appropriate physical and chemical properties used to treat poorly healing wounds. In addition, it will make it possible to design devices precisely controlling wound mechanics and to individualize a therapy depending on the type, size, and anatomical location of the wound in specific patients, which will increase the clinical efficiency of the therapy. Linking mechanobiology with the science of biomaterials and nanotechnology will enable in the near future precise interference in abnormal cell signaling responsible for the proliferation, differentiation, cell death, and restoration of the biological balance. The objective of this study is to point to the importance of mechanobiology in regeneration of skin damage and wound healing. The study describes the influence of rigidity of extracellular matrix and special restrictions on cell physiology. The study also defines how and what mechanical changes influence tissue regeneration and wound healing. The influence of mechanical signals in the process of proliferation, differentiation, and skin regeneration is tagged in the study.
Collapse
|
34
|
Growth Hormone-Releasing Peptide 6 Enhances the Healing Process and Improves the Esthetic Outcome of the Wounds. PLASTIC SURGERY INTERNATIONAL 2016; 2016:4361702. [PMID: 27200188 PMCID: PMC4854984 DOI: 10.1155/2016/4361702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/28/2016] [Indexed: 01/25/2023]
Abstract
In addition to its cytoprotective effects, growth hormone-releasing peptide 6 (GHRP-6) proved to reduce liver fibrotic induration. CD36 as one of the GHRP-6 receptors appears abundantly represented in cutaneous wounds granulation tissue. The healing response in a scenario of CD36 agonistic stimulation had not been previously investigated. Excisional full-thickness wounds (6 mmØ) were created in the dorsum of Wistar rats and topically treated twice a day for 5 days. The universal model of rabbit's ears hypertrophic scars was implemented and the animals were treated daily for 30 days. Treatments for both species were based on a CMC jelly composition containing GHRP-6 400 μg/mL. Wounds response characterization included closure dynamic, RT-PCR transcriptional profile, histology, and histomorphometric procedures. The rats experiment indicated that GHRP-6 pharmacodynamics involves attenuation of immunoinflammatory mediators, their effector cells, and the reduction of the expression of fibrotic cytokines. Importantly, in the hypertrophic scars rabbit's model, GHRP-6 intervention dramatically reduced the onset of exuberant scars by activating PPARγ and reducing the expression of fibrogenic cytokines. GHRP-6 showed no effect on the reversion of consolidated lesions. This evidence supports the notion that CD36 is an active and pharmacologically approachable receptor to attenuate wound inflammation and accelerate its closure so as to improve wound esthetic.
Collapse
|
35
|
Castleberry SA, Golberg A, Sharkh MA, Khan S, Almquist BD, Austen WG, Yarmush ML, Hammond PT. Nanolayered siRNA delivery platforms for local silencing of CTGF reduce cutaneous scar contraction in third-degree burns. Biomaterials 2016; 95:22-34. [PMID: 27108403 DOI: 10.1016/j.biomaterials.2016.04.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 04/06/2016] [Accepted: 04/10/2016] [Indexed: 01/12/2023]
Abstract
Wound healing is an incredibly complex biological process that often results in thickened collagen-enriched healed tissue called scar. Cutaneous scars lack many functional structures of the skin such as hair follicles, sweat glands, and papillae. The absence of these structures contributes to a number of the long-term morbidities of wound healing, including loss of function for tissues, increased risk of re-injury, and aesthetic complications. Scar formation is a pervasive factor in our daily lives; however, in the case of serious traumatic injury, scars can create long-lasting complications due to contraction and poor tissue remodeling. Within this report we target the expression of connective tissue growth factor (CTGF), a key mediator of TGFβ pro-fibrotic response in cutaneous wound healing, with controlled local delivery of RNA interference. Through this work we describe both a thorough in vitro analysis of nanolayer coated sutures for the controlled delivery of siRNA and its application to improve scar outcomes in a third-degree burn induced scar model in rats. We demonstrate that the knockdown of CTGF significantly altered the local expression of αSMA, TIMP1, and Col1a1, which are known to play roles in scar formation. The knockdown of CTGF within the healing burn wounds resulted in improved tissue remodeling, reduced scar contraction, and the regeneration of papillary structures within the healing tissue. This work adds support to a number of previous reports that indicate CTGF as a potential therapeutic target for fibrosis. Additionally, we believe that the controlled local delivery of siRNA from ultrathin polymer coatings described within this work is a promising approach in RNA interference that could be applied in developing improved cancer therapies, regenerative medicine, and fundamental scientific research.
Collapse
Affiliation(s)
- Steven A Castleberry
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, 02139, USA
| | - Alexander Golberg
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and the Shriners Burns Hospital, Boston, MA, 02114, USA; Porter School of Environmental Studies, Tel Aviv University, Ramat-Aviv, Tel Aviv, Israel
| | - Malak Abu Sharkh
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Saiqa Khan
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Benjamin D Almquist
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - William G Austen
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Martin L Yarmush
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and the Shriners Burns Hospital, Boston, MA, 02114, USA; Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08901, USA
| | - Paula T Hammond
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
36
|
Zhu Z, Ding J, Tredget EE. The molecular basis of hypertrophic scars. BURNS & TRAUMA 2016; 4:2. [PMID: 27574672 PMCID: PMC4963951 DOI: 10.1186/s41038-015-0026-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 12/30/2015] [Indexed: 02/05/2023]
Abstract
Hypertrophic scars (HTS) are caused by dermal injuries such as trauma and burns to the deep dermis, which are red, raised, itchy and painful. They can cause cosmetic disfigurement or contractures if craniofacial areas or mobile region of the skin are affected. Abnormal wound healing with more extracellular matrix deposition than degradation will result in HTS formation. This review will introduce the physiology of wound healing, dermal HTS formation, treatment and difference with keloids in the skin, and it also review the current advance of molecular basis of HTS including the involvement of cytokines, growth factors, and macrophages via chemokine pathway, to bring insights for future prevention and treatment of HTS.
Collapse
Affiliation(s)
- Zhensen Zhu
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Alberta Canada
- Department of Burn and Reconstructive Surgery, 2nd Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong China
| | - Jie Ding
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Alberta Canada
| | - Edward E. Tredget
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Alberta Canada
- Division of Plastic Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta Canada
| |
Collapse
|
37
|
Alteration of Connective Tissue Growth Factor (CTGF) Expression in Orbital Fibroblasts from Patients with Graves' Ophthalmopathy. PLoS One 2015; 10:e0143514. [PMID: 26599235 PMCID: PMC4657967 DOI: 10.1371/journal.pone.0143514] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 11/05/2015] [Indexed: 02/07/2023] Open
Abstract
Graves’ ophthalmopathy (GO) is a disfiguring and sometimes blinding disease, which is characterized by inflammation and swelling of orbital tissues, with fibrosis and adipogenesis being predominant features. The aim of this study is to investigate whether the expression levels of fibrosis-related genes, especially that of connective tissue growth factor (CTGF), are altered in orbital fibroblasts of patients with GO. The role of oxidative stress in the regulation of CTGF expression in GO orbital fibroblasts is also examined. By a SYBR Green-based real time quantitative PCR (RT-QPCR), we demonstrated that the mRNA expression levels of fibronectin, apolipoprotein J, and CTGF in cultured orbital fibroblasts from patients with GO were significantly higher than those of age-matched normal controls (p = 0.007, 0.037, and 0.002, respectively). In addition, the protein expression levels of fibronectin, apolipoprotein J, and CTGF analyzed by Western blot were also significantly higher in GO orbital fibroblasts (p = 0.046, 0.032, and 0.008, respectively) as compared with the control. Furthermore, after treatment of orbital fibroblasts with a sub-lethal dose of hydrogen peroxide (200 μM H2O2), we found that the H2O2-induced increase of CTGF expression was more pronounced in the GO orbital fibroblasts as compared with those in normal controls (20% vs. 7%, p = 0.007). Importantly, pre-incubation with antioxidants including N-acetylcysteine (NAC) and vitamin C, respectively, resulted in significant attenuation of the induction of CTGF in GO orbital fibroblasts in response to H2O2 (p = 0.004 and 0.015, respectively). Taken together, we suggest that oxidative stress plays a role in the alteration of the expression of CTGF in GO orbital fibroblasts that may contribute to the pathogenesis and progression of GO. Antioxidants may be used in combination with the therapeutic agents for effective treatment of GO.
Collapse
|
38
|
Aoki S, Takezawa T, Ikeda S, Narisawa Y, Oshikata-Miyazaki A, Miyauchi S, Hirayama H, Sawaguchi T, Chimuro T, Toda S. A new cell-free bandage-type artificial skin for cutaneous wounds. Wound Repair Regen 2015; 23:819-29. [DOI: 10.1111/wrr.12321] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 06/01/2015] [Indexed: 12/29/2022]
Affiliation(s)
- Shigehisa Aoki
- Department of Pathology and Microbiology; Faculty of Medicine, Saga University; Saga Japan
| | - Toshiaki Takezawa
- Division of Animal Sciences; National Institute of Agrobiological Sciences; Ibaraki Japan
| | - Satoshi Ikeda
- Department of Pathology and Microbiology; Faculty of Medicine, Saga University; Saga Japan
| | - Yutaka Narisawa
- Department of Dermatology, Faculty of Medicine; Saga University; Saga Japan
| | | | - Syohei Miyauchi
- Research and Development, Yutoku Pharmaceutical Industries Co. Ltd.; Saga Japan
| | - Hiroshi Hirayama
- Business Development; Yutoku Pharmaceutical Industries Co. Ltd.; Saga Japan
| | - Tomoya Sawaguchi
- Isehara Research Laboratory; Kanto Chemical Co. Inc.; Kanagawa Japan
| | - Tomoyuki Chimuro
- Isehara Research Laboratory; Kanto Chemical Co. Inc.; Kanagawa Japan
| | - Shuji Toda
- Department of Pathology and Microbiology; Faculty of Medicine, Saga University; Saga Japan
| |
Collapse
|
39
|
Loiselle AE, Yukata K, Geary MB, Kondabolu S, Shi S, Jonason JH, Awad HA, O’Keefe RJ. Development of antisense oligonucleotide (ASO) technology against Tgf-β signaling to prevent scarring during flexor tendon repair. J Orthop Res 2015; 33:859-66. [PMID: 25761254 PMCID: PMC4416995 DOI: 10.1002/jor.22890] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 03/01/2015] [Indexed: 02/04/2023]
Abstract
Flexor tendons (FT) in the hand provide near frictionless gliding to facilitate hand function. Upon injury and surgical repair, satisfactory healing is hampered by fibrous adhesions between the tendon and synovial sheath. In the present study we used antisense oligonucleotides (ASOs), specifically targeted to components of Tgf-β signaling, including Tgf-β1, Smad3 and Ctgf, to test the hypothesis that local delivery of ASOs and suppression of Tgf-β1 signaling would enhance murine FT healing by suppressing adhesion formation while maintaining strength. ASOs were injected in to the FT repair site at 2, 6 and 12 days post-surgery. ASO treatment suppressed target gene expression through 21 days. Treatment with Tgf-β1, Smad3 or Ctgf ASOs resulted in significant improvement in tendon gliding function at 14 and 21 days, relative to control. Consistent with a decrease in adhesions, Col3a1 expression was significantly decreased in Tgf-β1, Smad3 and Ctgf ASO treated tendons relative to control. Smad3 ASO treatment enhanced the maximum load at failure of healing tendons at 14 days, relative to control. Taken together, these data support the use of ASO treatment to improve FT repair, and suggest that modulation of the Tgf-β1 signaling pathway can reduce adhesions while maintaining the strength of the repair.
Collapse
Affiliation(s)
- Alayna E. Loiselle
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Kiminori Yukata
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Michael B. Geary
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Sirish Kondabolu
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Shanshan Shi
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Jennifer H. Jonason
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Hani A. Awad
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642,Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627
| | - Regis J. O’Keefe
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642,Department of Orthopaedic Surgery, Washington University in St. Louis School of Medicine, St. Louis, Missouri 63110,Corresponding Author: Regis O’Keefe, Department of Orthopaedic Surgery, Washington University in St. Louis School of Medicine, 660 South Euclid Ave., St. Louis, MO 63110-1093,
| |
Collapse
|
40
|
Xu H, Liu C, Sun Z, Guo X, Zhang Y, Liu M, Li P. CCN5 attenuates profibrotic phenotypes of fibroblasts through the Smad6-CCN2 pathway: Potential role in epidural fibrosis. Int J Mol Med 2015; 36:123-9. [PMID: 25901787 PMCID: PMC4494601 DOI: 10.3892/ijmm.2015.2190] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 03/27/2015] [Indexed: 12/31/2022] Open
Abstract
Epidural fibrosis is characterized by the development of dense and thick scar tissue adjacent to the dural mater and ranked as the major contributor for post-operative pain recurrence after laminectomy or discectomy. Recently, CCN5 exhibited an inhibitory effect on connective tissue growth factor (CTGF)/CCN2 (a critical regulator for fibrotic disease)-mediated fibrogenesis. However, its function in epidural fibrosis and the underlying mechanisms involved remain to be determined. In this study, an obvious downregulation of CCN5 was observed in scar tissues from laminectomized rats, concomitant with a marked upregulation of CCN2, suggesting a potential negative regulatory role of CCN5 in fibrogenesis. Furthermore, CCN5 overexpression notably mitigated transforming growth factor-β1-enhanced fibroblast viability and proliferation. Of note, CCN5 upregulation inhibited the switch of fibroblasts into myofibroblasts as its overexpression abrogated the expression of the myofibroblast marker, α-smooth muscle actin (α-SMA). CCN5 upregulation also reduced an increase in collagen type I, α1 (COL1A1) and total collagen concentrations. Additionally, CCN5 over expression decreased CCN2 expression and increased Smad6 phosphorylation. Mechanism analysis revealed that blocking Smad6 signaling significantly ameliorated the inhibitory effect of CCN5 on the CCN2 levels, accompanied by the reduction in cell proliferation and collagen production. These results confirm that CCN5 exerts an anti-fibrotic function by regulating the Smad6-CCN2 pathway, thereby indicating a potential approach for ameliorating epidural fibrosis after laminectomy.
Collapse
Affiliation(s)
- Honghai Xu
- Department of Orthopaedics, The Third Affiliated Hospital (Shaanxi Provincial People's Hospital), Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Cong Liu
- Xi'an Medical College, Xi'an, Shaanxi 710061, P.R. China
| | - Zhengming Sun
- Department of Orthopaedics, The Third Affiliated Hospital (Shaanxi Provincial People's Hospital), Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiong Guo
- Department of Public Health, Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yuelin Zhang
- Department of Neurosurgery, The Third Affiliated Hospital (Shaanxi Provincial People's Hospital), Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Mengting Liu
- Xi'an Medical College, Xi'an, Shaanxi 710061, P.R. China
| | - Peng Li
- Xi'an Medical College, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
41
|
Xu W, Hong SJ, Zeitchek M, Cooper G, Jia S, Xie P, Qureshi HA, Zhong A, Porterfield MD, Galiano RD, Surmeier DJ, Mustoe TA. Hydration status regulates sodium flux and inflammatory pathways through epithelial sodium channel (ENaC) in the skin. J Invest Dermatol 2014; 135:796-806. [PMID: 25371970 DOI: 10.1038/jid.2014.477] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 09/22/2014] [Accepted: 10/05/2014] [Indexed: 02/03/2023]
Abstract
Although it is known that the inflammatory response that results from disruption of epithelial barrier function after injury results in excessive scarring, the upstream signals remain unknown. It has also been observed that epithelial disruption results in reduced hydration status and that the use of occlusive dressings that prevent water loss from wounds decreases scar formation. We hypothesized that hydration status changes sodium homeostasis and induces sodium flux in keratinocytes, which result in activation of pathways responsible for keratinocyte-fibroblast signaling and ultimately lead to activation of fibroblasts. Here, we demonstrate that perturbations in epithelial barrier function lead to increased sodium flux in keratinocytes. We identified that sodium flux in keratinocytes is mediated by epithelial sodium channels (ENaCs) and causes increased secretion of proinflammatory cytokines, which activate fibroblast via the cyclooxygenase 2 (COX-2)/prostaglandin E2 (PGE2) pathway. Similar changes in signal transduction and sodium flux occur by increased sodium concentration, which simulates reduced hydration, in the media in epithelial cultures or human ex vivo skin cultures. Blockade of ENaC, prostaglandin synthesis, or PGE2 receptors all reduce markers of fibroblast activation and collagen synthesis. In addition, employing a validated in vivo excessive scar model in the rabbit ear, we demonstrate that utilization of either an ENaC blocker or a COX-2 inhibitor results in a marked reduction in scarring. Other experiments demonstrate that the activation of COX-2 in response to increased sodium flux is mediated through the PIK3/Akt pathway. Our results indicate that ENaC responds to small changes in sodium concentration with inflammatory mediators and suggest that the ENaC pathway is a potential target for a strategy to prevent fibrosis.
Collapse
Affiliation(s)
- Wei Xu
- Laboratory for Wound Repair and Regenerative Medicine, Department of Surgery/Plastic Surgery Division, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Seok Jong Hong
- Laboratory for Wound Repair and Regenerative Medicine, Department of Surgery/Plastic Surgery Division, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Michael Zeitchek
- Department of Agricultural and Biological Engineering, Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Garry Cooper
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Shengxian Jia
- Laboratory for Wound Repair and Regenerative Medicine, Department of Surgery/Plastic Surgery Division, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ping Xie
- Laboratory for Wound Repair and Regenerative Medicine, Department of Surgery/Plastic Surgery Division, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hannan A Qureshi
- Laboratory for Wound Repair and Regenerative Medicine, Department of Surgery/Plastic Surgery Division, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Aimei Zhong
- Laboratory for Wound Repair and Regenerative Medicine, Department of Surgery/Plastic Surgery Division, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Marshall D Porterfield
- Department of Agricultural and Biological Engineering, Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Robert D Galiano
- Laboratory for Wound Repair and Regenerative Medicine, Department of Surgery/Plastic Surgery Division, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - D James Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Thomas A Mustoe
- Laboratory for Wound Repair and Regenerative Medicine, Department of Surgery/Plastic Surgery Division, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.
| |
Collapse
|
42
|
Falke LL, Goldschmeding R, Nguyen TQ. A perspective on anti-CCN2 therapy for chronic kidney disease. Nephrol Dial Transplant 2014; 29 Suppl 1:i30-i37. [PMID: 24493868 DOI: 10.1093/ndt/gft430] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Kidney fibrosis is the common end point of chronic kidney disease independent of aetiology. Currently, no effective therapy exists to reduce kidney fibrosis. CCN2 appears to be an interesting candidate for anti-fibrotic drug targeting, because it holds a central position in the development of kidney fibrosis and interacts with a variety of factors that are involved in the fibrotic response, including transforming growth factor (TGF) β and Bone morphogenetic proteins. Although CCN2 modifies many pathways, it does not appear to have a membrane receptor of its own. Numerous experimental and clinical studies lowering CCN2 bioavailability have shown promising results with minimal adverse side effects. This review aims to provide an overview of the current state of CCN2 research with a focus on anti-fibrotic therapy.
Collapse
Affiliation(s)
- Lucas L Falke
- Department of Pathology, UMC Utrecht, Utrecht, Netherlands
| | | | | |
Collapse
|
43
|
Liu S, Thompson K, Leask A. CCN2 expression by fibroblasts is not required for cutaneous tissue repair. Wound Repair Regen 2014; 22:119-24. [PMID: 24393160 DOI: 10.1111/wrr.12131] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 10/10/2013] [Indexed: 12/17/2022]
Abstract
The CCN family of matricellular proteins, which includes CCN2 and CCN1, is believed to have a major in vivo role in controlling tissue morphogenesis and repair. In adult skin, the proadhesive matricellular protein connective tissue growth factor (CTGF/CCN2) is specifically up-regulated in fibrosis and wound healing. In mice, CCN2 is required for dermal fibrogenesis, but whether CCN2 is required for cutaneous tissue repair is unknown. To address this question, in this report we subjected adult mice bearing a fibroblast-specific deletion of CCN2 to the dermal punch model of cutaneous tissue repair. Loss of CCN2 did not appreciably affect the kinetics of tissue repair, collagen content, or the number of α-smooth muscle actin-positive cells. CCN1 (cyr61), which has in vitro effect similar to CCN2, is also induced in cutaneous tissue repair. Fibroblast-specific CCN1/CCN2 double knockout mice were also generated; loss of both CCN1 and CCN2 together did not appreciably affect cutaneous tissue repair. However, loss of CCN2 resulted in impaired recruitment of NG2-positive pericyte-like cells to the wound area. Collectively, these results indicate that neither CCN2 nor CCN1 is essential for cutaneous tissue repair; CCN2 appears to be required for recruitment of pericyte-like cells and may represent a specific antifibrotic target.
Collapse
Affiliation(s)
- Shangxi Liu
- Department of Dentistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | | | | |
Collapse
|
44
|
Hu X, Li N, Tao K, Fang X, Liu J, Wang Y, Wang H, Shi J, Wang Y, Ji P, Cai W, Bai X, Zhu X, Han J, Hu D. Effects of integrin ανβ3 on differentiation and collagen synthesis induced by connective tissue growth factor in human hypertrophic scar fibroblasts. Int J Mol Med 2014; 34:1323-34. [PMID: 25174803 DOI: 10.3892/ijmm.2014.1912] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 08/12/2014] [Indexed: 11/05/2022] Open
Abstract
CCN2 is a matricellular protein that appears to be important in scar formation. CCN2 mediates the pro-fibrotic effects in hypertrophic scars (HTSs) through an unknown mechanism. However, many activities of CCN2 protein are known to be mediated by direct binding to integrin receptors. In this study, we investigated the role of integrin α(ν)β(3) in the differentiation of hypertrophic scar fibroblasts (HTSFs) induced by CCN2. The levels of integrin α(ν)β(3) between normal skin and hypertrophic scar (HTS) tissues were compared, and integrin α(ν)β(3) was found to be upregulated in HTS. CCN2 was shown to induce HTSF differentiation and collagen (COL) synthesis at the mRNA and protein levels. Based on these results, the expression of integrin α(ν)β(3) was upregulated by CCN2 stimulation during HTSF differentiation. Blockade of integrin α(ν)β(3) prevented CCN2-induced HTSF differentiation and COL synthesis. Furthermore, the CCN2-induced increase in contractility of the HTSF in COL lattices was inhibited by integrin α(ν)β(3) blocking antibodies. HTSs were established in a rabbit ear model, and the inhibitor of integrin α(ν)β(3) significantly improved the architecture of the rabbit ear scar. Results of the present study showed that integrin α(ν)β(3) contributes to pro-fibrotic CCN2 signaling. Blocking this pathway may therefore be beneficial for the treatment of HTS.
Collapse
Affiliation(s)
- Xiaolong Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Na Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Ke Tao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xiaobing Fang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jiaqi Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yaojun Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Hongtao Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jihong Shi
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yunchuan Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Peng Ji
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Weixia Cai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xiaozhi Bai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xiongxiang Zhu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Juntao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
45
|
Rettinger CL, Fourcaudot AB, Hong SJ, Mustoe TA, Hale RG, Leung KP. In vitro characterization of scaffold-free three-dimensional mesenchymal stem cell aggregates. Cell Tissue Res 2014; 358:395-405. [PMID: 25012521 DOI: 10.1007/s00441-014-1939-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 06/03/2014] [Indexed: 01/14/2023]
Abstract
Mesenchymal stem cells (MSCs) are capable of self-renewal and differentiation along multiple cell lineages and have potential applications in a wide range of therapies. These cells are commonly cultured as monolayers on tissue culture plastic but possibly lose their cell-specific properties with time in vitro. There is growing interest in culturing adherent cells via three-dimensional (3D) techniques in order to recapitulate 3D in vivo conditions. We describe a novel method for generating and culturing rabbit MSCs as scaffold-free 3D cell aggregates by using micropatterned wells via a forced aggregation technique. The viability and proliferative capability of MSC aggregates were assessed via Live/Dead staining and 5-ethynyl-2'-deoxyuridine (EdU) incorporation. Enzyme-linked immunosorbent assay and antibody-based multiplex protein assays were used to quantify released growth factors and chemokines. The gene expression profile of MSCs as 3D aggregates relative to MSCs grown as monolayers was evaluated via quantitative real-time polymerase chain reaction. The rabbit MSCs were able to form compact cell aggregates and remained viable in 3D culture for up to 7 days. We also demonstrated enhanced gene and protein expression related to angiogenesis and wound healing in MSCs cultured under 3D conditions. In vitro tube formation and scratch assay revealed superior neovessel formation and greater cell recovery and migration in response to 3D conditioned media after wounding. Our data further suggest that adipose-derived stem cell aggregates have greater potential than dermal fibroblasts or bone-marrow-derived MSCs in accelerating wound healing and reducing scarring.
Collapse
Affiliation(s)
- Christina L Rettinger
- Dental and Trauma Research Detachment, United States Army Institute of Surgical Research, 3650 Chambers Pass, Building 3610, Fort Sam Houston, TX, 78234, USA
| | | | | | | | | | | |
Collapse
|
46
|
Ren Z, Hou Y, Ma S, Tao Y, Li J, Cao H, Ji L. Effects of CCN3 on fibroblast proliferation, apoptosis and extracellular matrix production. Int J Mol Med 2014; 33:1607-12. [PMID: 24715059 DOI: 10.3892/ijmm.2014.1735] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 03/14/2014] [Indexed: 11/06/2022] Open
Abstract
CCN2 and CCN3 belong to the CCN family of proteins, which show a high level of structural similarity.Previous studies have shown that CCN2 mediates the ability of transforming growth factor (TGF)‑β to stimulate collagen synthesis, leading to keloid formation. CCN2 and CCN3 are opposing factors in regulating the promoter activity and secretion of this extracellular matrix (ECM) protein. Thus, we hypothesize that CCN3 possesses an anti‑scarring effect. However, the exact mechanism of CCN3 in this anti‑scarring effect remains unclear. The aim of this study was to investigate the mechanism of CCN3 in reducing scar formation. Palatal fibroblasts were obtained from the explants of the oral palatal mucosa of 8‑week‑old male Sprague‑Dawley rats. CCN3 overexpression vector was constructed and then transfected into cells. The inhibitory effects of CCN3 on cell growth were detected via the 3‑(4,5‑dimethylthiazol‑2‑yl)‑2,5‑diphenyltetrazolium bromide (MTT) assay. Apoptosis was measured using an Annexin V‑fluorescein isothiocyanate (FITC)/propidium iodide (PI) apoptosis detection kit and flow cytometry. The expression levels of collagen I, collagen III and α‑smooth muscle actin (α‑SMA) were determined by western blot analysis and RT‑PCR. Following treatment with TGF‑β1, we detected the expression of CCN3 and Smad1 in the fibroblasts. CCN3 significantly inhibited the growth and induction of apoptosis of fibroblasts. The expression of collagen I, collagen III and α‑SMA was lower in the CCN3‑transfected group as compared to the control and vector groups. TGF‑β1 stimulation efficiently suppressed the expression of CCN3 at the mRNA and protein levels, and CCN3 was required for TGF‑β1‑induced Smad1 phosphorylation. Results of this study demonstrated that CCN3 is involved in the proliferation and apoptosis of fibroblasts and the synthesis of ECM proteins. Therefore, CCN3 may play an important role in the development of scar tissue, and may represent a novel therapeutic target for reducing scar formation.
Collapse
Affiliation(s)
- Zhanping Ren
- Department of Cranio‑Maxillofacial Trauma Plastic Surgery, Stomatology Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710004, P.R. China
| | - Yuxia Hou
- Department of Orthodontics, Stomatology Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710004, P.R. China
| | - Siwei Ma
- Department of Cranio‑Maxillofacial Trauma Plastic Surgery, Stomatology Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710004, P.R. China
| | - Yongwei Tao
- Department of Cranio‑Maxillofacial Trauma Plastic Surgery, Stomatology Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710004, P.R. China
| | - Jinfeng Li
- Department of Cranio‑Maxillofacial Trauma Plastic Surgery, Stomatology Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710004, P.R. China
| | - Huiqin Cao
- Department of Cranio‑Maxillofacial Trauma Plastic Surgery, Stomatology Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710004, P.R. China
| | - Lingling Ji
- Department of Orthodontics, Stomatology Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
47
|
Yang D, Gao YH, Tan KB, Zuo ZX, Yang WX, Hua X, Li PJ, Zhang Y, Wang G. Inhibition of hepatic fibrosis with artificial microRNA using ultrasound and cationic liposome-bearing microbubbles. Gene Ther 2013; 20:1140-8. [PMID: 23966015 DOI: 10.1038/gt.2013.41] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 06/10/2013] [Accepted: 06/12/2013] [Indexed: 12/17/2022]
Abstract
We sought to investigate the antifibrotic effects of an artificial microRNA (miRNA) targeting connective tissue growth factor (CTGF) using the ultrasound-targeted cationic liposome-bearing microbubble destruction gene delivery system. Cationic liposomes were conjugated with microbubbles using a biotin-avidin system. Plasmids carrying the most effective artificial miRNA sequences were delivered by ultrasound-targeted cationic liposome-bearing microbubble destruction gene delivery system to rats with hepatic fibrosis. The results show that this method of gene delivery effectively transported the plasmids to the rat liver. The artificial miRNA reduced hepatic fibrosis pathological alterations as well as the protein and mRNA expressions of CTGF and transforming growth factor β1. Furthermore, the CTGF gene silencing decreased the levels of type I collagen and α-smooth muscle actin (P<0.01). These data suggest that delivery of an artificial miRNA targeted against CTGF using ultrasound-targeted cationic liposome-bearing microbubble destruction may be an efficacious therapeutic method to ameliorate hepatic fibrosis.
Collapse
Affiliation(s)
- D Yang
- 1] Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China [2] Department of Ultrasound, 324th Military Hospital, Chongqing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Lee MJ, Byun MR, Furutani-Seiki M, Hong JH, Jung HS. YAP and TAZ regulate skin wound healing. J Invest Dermatol 2013; 134:518-525. [PMID: 24108406 DOI: 10.1038/jid.2013.339] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 07/10/2013] [Accepted: 07/13/2013] [Indexed: 11/09/2022]
Abstract
The Hippo signaling pathway regulates organ size, tissue regeneration, and stem cell self-renewal. The two key downstream transcription coactivators in this pathway, Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), mediate the major gene regulation and biological functions of the Hippo pathway. The biological functions of YAP and TAZ in many tissues are known; however, their roles in skin wound healing remain unclear. To analyze whether YAP and/or TAZ are required for cutaneous wound healing, we performed small interfering RNA (siRNA)-mediated knockdown of YAP/TAZ in full-thickness skin wounds. YAP is strongly expressed in the nucleus and cytoplasm in the epidermis and hair follicle. Interestingly, YAP is expressed in the nucleus in the dermis at 2 and 7 days after wounding. TAZ normally localizes to the cytoplasm in the dermis but is distributed in both the nucleus and cytoplasm at 1 day after wounding. The knockdown of YAP and TAZ markedly delayed the rate of wound closure and reduced the transforming growth factor-β1 (TGF-β1) expression in the wound. YAP and TAZ also modulate the expression of TGF-β1 signaling pathway components such as Smad-2, p21, and Smad-7. These results suggest that YAP and TAZ localization to the nucleus is required for skin wound healing.
Collapse
Affiliation(s)
- Min-Jung Lee
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Brain Korea 21 Project, Oral Science Research Center, College of Dentistry, Yonsei Center of Biotechnology, Yonsei University, Seoul, Korea
| | - Mi Ran Byun
- School of Life Science and Biotechnology, Korea University, Seoul, Korea
| | | | - Jeong-Ho Hong
- School of Life Science and Biotechnology, Korea University, Seoul, Korea
| | - Han-Sung Jung
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Brain Korea 21 Project, Oral Science Research Center, College of Dentistry, Yonsei Center of Biotechnology, Yonsei University, Seoul, Korea; Oral Biosciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR.
| |
Collapse
|
49
|
Glim JE, van Egmond M, Niessen FB, Everts V, Beelen RHJ. Detrimental dermal wound healing: what can we learn from the oral mucosa? Wound Repair Regen 2013; 21:648-60. [PMID: 23927738 DOI: 10.1111/wrr.12072] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Accepted: 06/01/2013] [Indexed: 12/11/2022]
Abstract
Wounds in adults are frequently accompanied by scar formation. This scar can become fibrotic due to an imbalance between extracellular matrix (ECM) synthesis and ECM degradation. Oral mucosal wounds, however, heal in an accelerated fashion, displaying minimal scar formation. The exact mechanisms of scarless oral healing are yet to be revealed. This review highlights possible mechanisms involved in the difference between scar-forming dermal vs. scarless oral mucosal wound healing. Differences were found in expression of ECM components, such as procollagen I and tenascin-C. Oral wounds contained fewer immune mediators, blood vessels, and profibrotic mediators but had more bone marrow-derived cells, a higher reepithelialization rate, and faster proliferation of fibroblasts compared with dermal wounds. These results form a basis for further research that should be focused on the relations among ECM, immune cells, growth factors, and fibroblast phenotypes, as understanding scarless oral mucosal healing may ultimately lead to novel therapeutic strategies to prevent fibrotic scars.
Collapse
Affiliation(s)
- Judith E Glim
- Department of Molecular Cell Biology & Immunology, VU University Medical Center, Amsterdam, The Netherlands; Department of Plastic and Reconstructive Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
50
|
Leask A. CCN2: a novel, specific and valid target for anti-fibrotic drug intervention. Expert Opin Ther Targets 2013; 17:1067-71. [PMID: 23848501 DOI: 10.1517/14728222.2013.812074] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Prior attempts at developing anti-fibrotic therapies have focused on using growth factors and cytokines as targets. However, growth factors and cytokines have effects on normal physiology as well as fibrosis, making effective drug development difficult. AREAS COVERED Matricellular proteins alter the cellular microenvironment and hence cellular signaling responses to cytokines and growth factors. A survey of Pubmed reveals that the expression pattern of matricellular proteins notably that of CCN2 (connective tissue growth factor) is often altered in pathophysiological conditions such as fibrosis. Moreover, data presented in recent publications suggests that CCN2 directly mediates fibrosis. EXPERT OPINION As a result of these features, matricellular proteins such as CCN2, a member of the CCN family of matricellular proteins, might be ideal targets against which to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Andrew Leask
- The University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Physiology & Pharmacology, Schulich Dentistry, London, Ontario, N6A 5C1, Canada.
| |
Collapse
|