1
|
Jin H, Yoon HJ, Jiang E, Liu J, Yoon HS, Choi JS, Moon J, Qi H, Yoon KC. Therapeutic Effects of Human Placental Extracts Eye Drops in Experimental Dry Eye and Alkali Burn. J Ocul Pharmacol Ther 2025; 41:141-149. [PMID: 39718797 DOI: 10.1089/jop.2024.0121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Abstract
Purpose: To evaluate the efficacy of human placental extract (HPE) eye drops compared to that of carboxymethylcellulose (CMC) and human peripheral blood serum (HPBS) eye drops in a mouse model of experimental dry eye (EDE) and corneal alkali burns. Methods: EDE and alkali burn models were induced in C57BL/6 mice using desiccating stress and NaOH, respectively. In both the EDE and alkali burn models, treatment groups received CMC, HPBS, or HPE eye drops. In EDE model, tear volume, tear break-up time (TBUT), and total corneal fluorescein staining score (CFSS) were measured. ROS were detected with 2',7'-dichlorodihydrofluorescein diacetate. Conjunctiva goblet cells were identified by periodic acid-Schiff staining, and corneal epithelial apoptosis was detected by TUNEL assay. In alkali burn model, the area and diameter of epithelial defects were assessed in each group. Results: In the EDE model, tear volume, CFSS, and epithelial apoptosis were significantly improved in all treatment groups. Compared to the CMC group, the HPE group showed a better improvement in the production of tear volume, TBUT, CFSS, ROS, and conjunctiva goblet cell density. There were no significant differences in parameters between the HPBS and HPE groups except for TBUT at 14 days. In the alkali burn model, the HPE group had a smaller area compared to the control and CMC groups and a shorter diameter compared to the control group. Conclusion: HPE eye drops were as effective as HPBS eye drops in improving the clinical signs and ocular surface oxidative damage of EDE and in promoting corneal epithelialization after alkali burn.
Collapse
Affiliation(s)
- Hui Jin
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju, Korea
- Department of Biomedical Sciences and Centers for Creative Biomedical Scientists at Chonnam National University, Gwangju, Korea
| | - Hyeon-Jeong Yoon
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Enying Jiang
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju, Korea
- Department of Biomedical Sciences and Centers for Creative Biomedical Scientists at Chonnam National University, Gwangju, Korea
| | - Jingting Liu
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Hee Su Yoon
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Ji Suk Choi
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Jayoung Moon
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Hong Qi
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
| | - Kyung Chul Yoon
- Department of Ophthalmology, Chonnam National University Medical School and Hospital, Gwangju, Korea
- Department of Biomedical Sciences and Centers for Creative Biomedical Scientists at Chonnam National University, Gwangju, Korea
| |
Collapse
|
2
|
Yu M, Wang S, Lin D. Mechanism and Application of Biomaterials Targeting Reactive Oxygen Species and Macrophages in Inflammation. Int J Mol Sci 2024; 26:245. [PMID: 39796102 PMCID: PMC11720555 DOI: 10.3390/ijms26010245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/22/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Inflammation, an adaptive reaction to harmful stimuli, is a necessary immune system response and can be either acute or chronic. Since acute inflammation tends to eliminate harmful stimuli and restore equilibrium, it is generally advantageous to the organism. Chronic inflammation, however, is caused by either increased inflammatory signaling or decreased pro-anti-inflammatory signaling. According to current studies, inflammation is thought to be a major factor in a number of chronic diseases, including diabetes, cancer, arthritis, inflammatory bowel disease, and obesity. Consequently, reducing inflammation is essential for both preventing and delaying diseases. The application of biomaterials in the treatment of inflammatory illnesses has grown in recent years. A variety of biomaterials can be implanted either by themselves or in conjunction with other bioactive ingredients and therapeutic agents. The mechanisms of action and therapeutic applications of well-known anti-inflammatory biomaterials are the main topics of this article.
Collapse
|
3
|
Giannasi C, Cadelano F, Della Morte E, Baserga C, Mazzucato C, Niada S, Baj A. Unlocking the Therapeutic Potential of Adipose-Derived Stem Cell Secretome in Oral and Maxillofacial Medicine: A Composition-Based Perspective. BIOLOGY 2024; 13:1016. [PMID: 39765683 PMCID: PMC11673083 DOI: 10.3390/biology13121016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 01/03/2025]
Abstract
The adipose-derived stem cell (ADSC) secretome is widely studied for its immunomodulatory and regenerative properties, yet its potential in maxillofacial medicine remains largely underexplored. This review takes a composition-driven approach, beginning with a list of chemokines, cytokines, receptors, and inflammatory and growth factors quantified in the ADSC secretome to infer its potential applications in this medical field. First, a review of the literature confirmed the presence of 107 bioactive factors in the secretome of ADSCs or other types of mesenchymal stem cells. This list was then analyzed using the Search Tool for Retrieval of Interacting Genes/Proteins (STRING) software, revealing 844 enriched biological processes. From these, key processes were categorized into three major clinical application areas: immunoregulation (73 factors), bone regeneration (13 factors), and wound healing and soft tissue regeneration (27 factors), with several factors relevant to more than one area. The most relevant molecules were discussed in the context of existing literature to explore their therapeutic potential based on available evidence. Among these, TGFB1, IL10, and CSF2 have been shown to modulate immune and inflammatory responses, while OPG, IL6, HGF, and TIMP1 contribute to bone regeneration and tissue repair. Although the ADSC secretome holds great promise in oral and maxillofacial medicine, further research is needed to optimize its application and validate its clinical efficacy.
Collapse
Affiliation(s)
- Chiara Giannasi
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20100 Milan, Italy; (F.C.); (A.B.)
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Francesca Cadelano
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20100 Milan, Italy; (F.C.); (A.B.)
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Elena Della Morte
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Camilla Baserga
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Camilla Mazzucato
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Stefania Niada
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Alessandro Baj
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20100 Milan, Italy; (F.C.); (A.B.)
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| |
Collapse
|
4
|
Kamal R, Awasthi A, Pundir M, Thakur S. Healing the diabetic wound: Unlocking the secrets of genes and pathways. Eur J Pharmacol 2024; 975:176645. [PMID: 38759707 DOI: 10.1016/j.ejphar.2024.176645] [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: 03/04/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Diabetic wounds (DWs) are open sores that can occur anywhere on a diabetic patient's body. They are often complicated by infections, hypoxia, oxidative stress, hyperglycemia, and reduced growth factors and nucleic acids. The healing process involves four phases: homeostasis, inflammation, proliferation, and remodeling, regulated by various cellular and molecular events. Numerous genes and signaling pathways such as VEGF, TGF-β, NF-κB, PPAR-γ, MMPs, IGF, FGF, PDGF, EGF, NOX, TLR, JAK-STAT, PI3K-Akt, MAPK, ERK, JNK, p38, Wnt/β-catenin, Hedgehog, Notch, Hippo, FAK, Integrin, and Src pathways are involved in these events. These pathways and genes are often dysregulated in DWs leading to impaired healing. The present review sheds light on the pathogenesis, healing process, signaling pathways, and genes involved in DW. Further, various therapeutic strategies that target these pathways and genes via nanotechnology are also discussed. Additionally, clinical trials on DW related to gene therapy are also covered in the present review.
Collapse
Affiliation(s)
- Raj Kamal
- Department of Quality Assurance, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Ankit Awasthi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| | - Mandeep Pundir
- School of Pharmaceutical Sciences, RIMT University, Punjab, 142001, India; Chitkara College of Pharmacy, Chitkara University, Punjab, 142001, India
| | - Shubham Thakur
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India
| |
Collapse
|
5
|
Pancheri NM, Ellingson AJ, Marchus CR, Durgesh V, Verhage T, Yensen N, Schiele NR. Lysyl Oxidase Production by Murine C3H10T1/2 Mesenchymal Stem Cells Is Increased by TGFβs and Differentially Modulated by Mechanical Stimuli. Stem Cells Dev 2024; 33:355-364. [PMID: 38770821 DOI: 10.1089/scd.2023.0295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
Tendons are frequently injured and have limited regenerative capacity. This motivates tissue engineering efforts aimed at restoring tendon function through strategies to direct functional tendon formation. Generation of a crosslinked collagen matrix is paramount to forming mechanically functional tendon. However, it is unknown how lysyl oxidase (LOX), the primary mediator of enzymatic collagen crosslinking, is regulated by stem cells. This study investigates how multiple factors previously identified to promote tendon formation and healing (transforming growth factor [TGF]β1 and TGFβ2, mechanical stimuli, and hypoxia-inducible factor [HIF]-1α) regulate LOX production in the murine C3H10T1/2 mesenchymal stem cell (MSC) line. We hypothesized that TGFβ signaling promotes LOX activity in C3H10T1/2 MSCs, which is regulated by both mechanical stimuli and HIF-1α activation. TGFβ1 and TGFβ2 increased LOX levels as a function of concentration and time. Inhibiting the TGFβ type I receptor (TGFβRI) decreased TGFβ2-induced LOX production by C3H10T1/2 MSCs. Low (5 mPa) and high (150 mPa) magnitudes of fluid shear stress were applied to test impacts of mechanical stimuli, but without TGFβ2, loading alone did not alter LOX levels. Low loading (5 mPa) with TGFβ2 increased LOX at 7 days greater than TGFβ2 treatment alone. Neither HIF-1α knockdown (siRNA) nor activation (CoCl2) affected LOX levels. Ultimately, results suggest that TGFβ2 and appropriate loading magnitudes contribute to LOX production by C3H10T1/2 MSCs. Potential application of these findings includes treatment with TGFβ2 and appropriate mechanical stimuli to modulate LOX production by stem cells to ultimately control collagen matrix stiffening and support functional tendon formation.
Collapse
Affiliation(s)
- Nicholas M Pancheri
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho, USA
| | - Allison J Ellingson
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho, USA
| | - Colin R Marchus
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho, USA
| | - Vibhav Durgesh
- Department of Mechanical Engineering, University of Idaho, Moscow, Idaho, USA
| | - Tabitha Verhage
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho, USA
| | - Nicholas Yensen
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho, USA
| | - Nathan R Schiele
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho, USA
| |
Collapse
|
6
|
Xiang Y, Zhao Y, Cheng T, Sun S, Wang J, Pei R. Implantable Neural Microelectrodes: How to Reduce Immune Response. ACS Biomater Sci Eng 2024; 10:2762-2783. [PMID: 38591141 DOI: 10.1021/acsbiomaterials.4c00238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Implantable neural microelectrodes exhibit the great ability to accurately capture the electrophysiological signals from individual neurons with exceptional submillisecond precision, holding tremendous potential for advancing brain science research, as well as offering promising avenues for neurological disease therapy. Although significant advancements have been made in the channel and density of implantable neural microelectrodes, challenges persist in extending the stable recording duration of these microelectrodes. The enduring stability of implanted electrode signals is primarily influenced by the chronic immune response triggered by the slight movement of the electrode within the neural tissue. The intensity of this immune response increases with a higher bending stiffness of the electrode. This Review thoroughly analyzes the sequential reactions evoked by implanted electrodes in the brain and highlights strategies aimed at mitigating chronic immune responses. Minimizing immune response mainly includes designing the microelectrode structure, selecting flexible materials, surface modification, and controlling drug release. The purpose of this paper is to provide valuable references and ideas for reducing the immune response of implantable neural microelectrodes and stimulate their further exploration in the field of brain science.
Collapse
Affiliation(s)
- Ying Xiang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yuewu Zhao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Tingting Cheng
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Shengkai Sun
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jine Wang
- Jiangxi Institute of Nanotechnology, Nanchang 330200, China
- College of Medicine and Nursing, Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, Dezhou University, Dezhou 253023, China
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| |
Collapse
|
7
|
Danielpour D. Advances and Challenges in Targeting TGF-β Isoforms for Therapeutic Intervention of Cancer: A Mechanism-Based Perspective. Pharmaceuticals (Basel) 2024; 17:533. [PMID: 38675493 PMCID: PMC11054419 DOI: 10.3390/ph17040533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
The TGF-β family is a group of 25 kDa secretory cytokines, in mammals consisting of three dimeric isoforms (TGF-βs 1, 2, and 3), each encoded on a separate gene with unique regulatory elements. Each isoform plays unique, diverse, and pivotal roles in cell growth, survival, immune response, and differentiation. However, many researchers in the TGF-β field often mistakenly assume a uniform functionality among all three isoforms. Although TGF-βs are essential for normal development and many cellular and physiological processes, their dysregulated expression contributes significantly to various diseases. Notably, they drive conditions like fibrosis and tumor metastasis/progression. To counter these pathologies, extensive efforts have been directed towards targeting TGF-βs, resulting in the development of a range of TGF-β inhibitors. Despite some clinical success, these agents have yet to reach their full potential in the treatment of cancers. A significant challenge rests in effectively targeting TGF-βs' pathological functions while preserving their physiological roles. Many existing approaches collectively target all three isoforms, failing to target just the specific deregulated ones. Additionally, most strategies tackle the entire TGF-β signaling pathway instead of focusing on disease-specific components or preferentially targeting tumors. This review gives a unique historical overview of the TGF-β field often missed in other reviews and provides a current landscape of TGF-β research, emphasizing isoform-specific functions and disease implications. The review then delves into ongoing therapeutic strategies in cancer, stressing the need for more tools that target specific isoforms and disease-related pathway components, advocating mechanism-based and refined approaches to enhance the effectiveness of TGF-β-targeted cancer therapies.
Collapse
Affiliation(s)
- David Danielpour
- Case Comprehensive Cancer Center Research Laboratories, The Division of General Medical Sciences-Oncology, Case Western Reserve University, Cleveland, OH 44106, USA; ; Tel.: +1-216-368-5670; Fax: +1-216-368-8919
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA
- Institute of Urology, University Hospitals, Cleveland, OH 44106, USA
| |
Collapse
|
8
|
Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C, He J. TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther 2024; 9:61. [PMID: 38514615 PMCID: PMC10958066 DOI: 10.1038/s41392-024-01764-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 08/31/2023] [Accepted: 01/31/2024] [Indexed: 03/23/2024] Open
Abstract
Transforming growth factor (TGF)-β is a multifunctional cytokine expressed by almost every tissue and cell type. The signal transduction of TGF-β can stimulate diverse cellular responses and is particularly critical to embryonic development, wound healing, tissue homeostasis, and immune homeostasis in health. The dysfunction of TGF-β can play key roles in many diseases, and numerous targeted therapies have been developed to rectify its pathogenic activity. In the past decades, a large number of studies on TGF-β signaling have been carried out, covering a broad spectrum of topics in health, disease, and therapeutics. Thus, a comprehensive overview of TGF-β signaling is required for a general picture of the studies in this field. In this review, we retrace the research history of TGF-β and introduce the molecular mechanisms regarding its biosynthesis, activation, and signal transduction. We also provide deep insights into the functions of TGF-β signaling in physiological conditions as well as in pathological processes. TGF-β-targeting therapies which have brought fresh hope to the treatment of relevant diseases are highlighted. Through the summary of previous knowledge and recent updates, this review aims to provide a systematic understanding of TGF-β signaling and to attract more attention and interest to this research area.
Collapse
Affiliation(s)
- Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
9
|
Choi AJ, Hefley BS, Nicholas SE, Cunningham RL, Karamichos D. Novel Correlation between TGF-β1/-β3 and Hormone Receptors in the Human Corneal Stroma. Int J Mol Sci 2023; 24:13635. [PMID: 37686439 PMCID: PMC10487450 DOI: 10.3390/ijms241713635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/22/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
This study investigated the interplay between transforming growth factor beta (TGF-β1/T1 and TGF-β3/T3), and sex hormone receptors using our 3D in vitro cornea stroma model. Primary human corneal fibroblasts (HCFs) from healthy donors were plated in transwells at 106 cells/well and cultured for four weeks. HCFs were supplemented with stable vitamin C (VitC) and stimulated with T1 or T3. 3D construct proteins were analyzed for the androgen receptor (AR), progesterone receptor (PR), estrogen receptor alpha (ERα) and beta (ERβ), luteinizing hormone receptor (LHR), follicle-stimulating hormone receptor (FSHR), gonadotropin-releasing hormone receptor (GnRHR), KiSS1-derived peptide receptor (KiSS1R/GPR54), and follicle-stimulating hormone subunit beta (FSH-B). In female constructs, T1 significantly upregulated AR, PR, ERα, FSHR, GnRHR, and KiSS1R. In male constructs, T1 significantly downregulated FSHR and FSH-B and significantly upregulated ERα, ERβ, and GnRHR. T3 caused significant upregulation in expressions PR, ERα, ERβ, LHR, FSHR, and GNRHR in female constructs, and significant downregulation of AR, ERα, and FSHR in male constructs. Semi-quantitative Western blot findings present the interplay between sex hormone receptors and TGF-β isoforms in the corneal stroma, which is influenced by sex as a biological variable (SABV). Additional studies are warranted to fully delineate their interactions and signaling mechanisms.
Collapse
Affiliation(s)
- Alexander J. Choi
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (A.J.C.); (B.S.H.); (S.E.N.)
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| | - Brenna S. Hefley
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (A.J.C.); (B.S.H.); (S.E.N.)
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| | - Sarah E. Nicholas
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (A.J.C.); (B.S.H.); (S.E.N.)
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| | - Rebecca L. Cunningham
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| | - Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (A.J.C.); (B.S.H.); (S.E.N.)
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
10
|
Rahman T, Das A, Abir MH, Nafiz IH, Mahmud AR, Sarker MR, Emran TB, Hassan MM. Cytokines and their role as immunotherapeutics and vaccine Adjuvants: The emerging concepts. Cytokine 2023; 169:156268. [PMID: 37320965 DOI: 10.1016/j.cyto.2023.156268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023]
Abstract
Cytokines are a protein family comprising interleukins, lymphokines, chemokines, monokines and interferons. They are significant constituents of the immune system, and they act in accordance with specific cytokine inhibiting compounds and receptors for the regulation of immune responses. Cytokine studies have resulted in the establishment of newer therapies which are being utilized for the treatment of several malignant diseases. The advancement of these therapies has occurred from two distinct strategies. The first strategy involves administrating the recombinant and purified cytokines, and the second strategy involves administrating the therapeutics which inhibits harmful effects of endogenous and overexpressed cytokines. Colony stimulating factors and interferons are two exemplary therapeutics of cytokines. An important effect of cytokine receptor antagonist is that they can serve as anti-inflammatory agents by altering the treatments of inflammation disorder, therefore inhibiting the effects of tumour necrosis factor. In this article, we have highlighted the research behind the establishment of cytokines as therapeutics and vaccine adjuvants, their role of immunotolerance, and their limitations.
Collapse
Affiliation(s)
- Tanjilur Rahman
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Ayan Das
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Mehedy Hasan Abir
- Faculty of Food Science and Technology, Chattogram Veterinary and Animal Sciences University, Chattogram 4225, Bangladesh
| | - Iqbal Hossain Nafiz
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Aar Rafi Mahmud
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Md Rifat Sarker
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chattogram 4381, Bangladesh; Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Mohammad Mahmudul Hassan
- Department of Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Chattogram 4225, Bangladesh; Queensland Alliance for One Health Sciences, School of Veterinary Science, The University of Queensland, Queensland 4343, Australia.
| |
Collapse
|
11
|
Chen PY, Qin L, Simons M. TGFβ signaling pathways in human health and disease. Front Mol Biosci 2023; 10:1113061. [PMID: 37325472 PMCID: PMC10267471 DOI: 10.3389/fmolb.2023.1113061] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/27/2023] [Indexed: 06/17/2023] Open
Abstract
Transforming growth factor beta (TGFβ) is named for the function it was originally discovered to perform-transformation of normal cells into aggressively growing malignant cells. It became apparent after more than 30 years of research, however, that TGFβ is a multifaceted molecule with a myriad of different activities. TGFβs are widely expressed with almost every cell in the human body producing one or another TGFβ family member and expressing its receptors. Importantly, specific effects of this growth factor family differ in different cell types and under different physiologic and pathologic conditions. One of the more important and critical TGFβ activities is the regulation of cell fate, especially in the vasculature, that will be the focus of this review.
Collapse
Affiliation(s)
- Pei-Yu Chen
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Lingfeng Qin
- Department of Surgery, Yale University School of Medicine, New Haven, CT, United States
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
12
|
Gallegos-Alcalá P, Jiménez M, Cervantes-García D, Córdova-Dávalos LE, Gonzalez-Curiel I, Salinas E. Glycomacropeptide Protects against Inflammation and Oxidative Stress, and Promotes Wound Healing in an Atopic Dermatitis Model of Human Keratinocytes. Foods 2023; 12:foods12101932. [PMID: 37238750 DOI: 10.3390/foods12101932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/04/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
Keratinocytes are actively implicated in the physiopathology of atopic dermatitis (AD), a skin allergy condition widely distributed worldwide. Glycomacropeptide (GMP) is a milk-derived bioactive peptide generated during cheese making processes or gastric digestion. It has antiallergic and skin barrier restoring properties when it is orally administered in experimental AD. This study aimed to evaluate the effect of GMP on the inflammatory, oxidative, proliferative, and migratory responses of HaCaT keratinocytes in an in vitro AD model. GMP protected keratinocytes from death and apoptosis in a dose dependent manner. GMP at 6.3 and 25 mg/mL, respectively, reduced nitric oxide by 50% and 83.2% as well as lipid hydroperoxides by 27.5% and 45.18% in activated HaCaT cells. The gene expression of TSLP, IL33, TARC, MDC, and NGF was significantly downregulated comparably to control by GMP treatment in activated keratinocytes, while that of cGRP was enhanced. Finally, in an AD microenvironment, GMP at 25 mg/mL stimulated HaCaT cell proliferation, while concentrations of 0.01 and 0.1 mg/mL promoted the HaCaT cell migration. Therefore, we demonstrate that GMP has anti-inflammatory and antioxidative properties and stimulates wound closure on an AD model of keratinocytes, which could support its reported bioactivity in vivo.
Collapse
Affiliation(s)
- Pamela Gallegos-Alcalá
- Laboratory of Immunology, Department of Microbiology, Center of Basic Science, Universidad Autónoma de Aguascalientes, Av. Universidad # 940, Aguascalientes 20100, Mexico
| | - Mariela Jiménez
- Laboratory of Immunology, Department of Microbiology, Center of Basic Science, Universidad Autónoma de Aguascalientes, Av. Universidad # 940, Aguascalientes 20100, Mexico
| | - Daniel Cervantes-García
- Laboratory of Immunology, Department of Microbiology, Center of Basic Science, Universidad Autónoma de Aguascalientes, Av. Universidad # 940, Aguascalientes 20100, Mexico
- National Council of Science and Technology, Av. de los Insurgentes Sur 1582, Crédito Constructor, Benito Juárez, Ciudad de México 03940, Mexico
| | - Laura Elena Córdova-Dávalos
- Laboratory of Immunology, Department of Microbiology, Center of Basic Science, Universidad Autónoma de Aguascalientes, Av. Universidad # 940, Aguascalientes 20100, Mexico
| | - Irma Gonzalez-Curiel
- Laboratory of Immunotoxicology and Experimental Therapeutics, Unidad Académica de Ciencias Químicas, Universidad Autónoma de Zacatecas, Carr. Zac.-Gdl. Km 6, Zacatecas 98160, Mexico
| | - Eva Salinas
- Laboratory of Immunology, Department of Microbiology, Center of Basic Science, Universidad Autónoma de Aguascalientes, Av. Universidad # 940, Aguascalientes 20100, Mexico
| |
Collapse
|
13
|
Kim HY, Im HY, Chang HK, Jeong HD, Park JH, Kim HI, Yi HS, Kim YS. Correlation between Collagen Type I/III Ratio and Scar Formation in Patients Undergoing Immediate Reconstruction with the Round Block Technique after Breast-Conserving Surgery. Biomedicines 2023; 11:biomedicines11041089. [PMID: 37189707 DOI: 10.3390/biomedicines11041089] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
The aim of this study was to investigate the relationship between the collagen type I/III ratio and scarring in patients who underwent immediate reconstruction with the round block technique (RBT) after breast conservation surgery. Seventy-eight patients were included, and demographic and clinical characteristics were recorded. The collagen type I/III ratio was measured using immunofluorescence staining and digital imaging, and scarring was assessed using the Vancouver Scar Scale (VSS). The mean VSS scores were 1.92 ± 2.01 and 1.79 ± 1.89, as assessed by two independent plastic surgeons, with good reliability of the scores. A significant positive correlation was found between VSS and the collagen type I/III ratio (r = 0.552, p < 0.01), and a significant negative correlation was found between VSS and the collagen type III content (r = −0.326, p < 0.05). Multiple linear regression analysis showed that the collagen type I/III ratio had a significant positive effect on VSS (β = 0.415, p = 0.028), whereas the collagen type I and collagen type III content had no significant effect on VSS. These findings suggest that the collagen type I/III ratio is associated with scar development in patients undergoing RBT after breast conservation surgery. Further research is needed to develop a patient-specific scar prediction model based on genetic factors affecting the collagen type I/III ratio.
Collapse
Affiliation(s)
- Hyo-young Kim
- We Are the Plastic Surgery, 415, Haeun-daero, Haeundae-gu, Busan 48064, Republic of Korea
| | - Ho-young Im
- Department of Plastic and Reconstructive Surgery, College of Medicine, Kosin University, 262, Seo-gu, Busan 49267, Republic of Korea
| | - Hee-kyung Chang
- Department of Pathology, College of Medicine, Kosin University, 262, Seo-gu, Busan 49267, Republic of Korea
| | - Hwan-do Jeong
- Kosin Innovative Smart Healthcare Research Center, Kosin University Gospel Hospital, 262, Seo-gu, Busan 49267, Republic of Korea
| | - Jin-hyung Park
- Department of Plastic and Reconstructive Surgery, College of Medicine, Kosin University, 262, Seo-gu, Busan 49267, Republic of Korea
| | - Hong-il Kim
- Department of Plastic and Reconstructive Surgery, College of Medicine, Kosin University, 262, Seo-gu, Busan 49267, Republic of Korea
| | - Hyung-suk Yi
- Department of Plastic and Reconstructive Surgery, College of Medicine, Kosin University, 262, Seo-gu, Busan 49267, Republic of Korea
| | - Yoon-soo Kim
- Department of Plastic and Reconstructive Surgery, College of Medicine, Kosin University, 262, Seo-gu, Busan 49267, Republic of Korea
| |
Collapse
|
14
|
Maier M, Olthoff S, Hill K, Zosel C, Magauer T, Wein LA, Schaefer M. KS0365, a novel activator of the transient receptor potential vanilloid 3 (TRPV3) channel, accelerates keratinocyte migration. Br J Pharmacol 2022; 179:5290-5304. [PMID: 35916168 DOI: 10.1111/bph.15937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 06/09/2022] [Accepted: 07/06/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Ca2+ signalling mediated by the thermosensitive, non-selective, Ca2+ -permeable transient receptor potential channel TRPV3 is assumed to play a critical role in regulating several aspects of skin functions, such as keratinocyte proliferation, differentiation, skin barrier formation and wound healing. Studying the function of TRPV3 in skin homeostasis, however, is still constrained by a lack of potent and selective pharmacological modulators of TRPV3. EXPERIMENTAL APPROACH By screening an in-house compound library using fluorometric intracellular Ca2+ assays, we identified two chemically related hits. The more potent and efficient TRPV3 activator KS0365 was further evaluated in fluo-4-assisted Ca2+ assays, different Ca2+ imaging approaches, electrophysiological studies, cytotoxicity and migration assays. KEY RESULTS KS0365 activated recombinant and native mouse TRPV3 more potently and with a higher efficacy compared to 2-APB and did not activate TRPV1, TRPV2 or TRPV4 channels. The activation of TRPV3 by KS0365 super-additively accelerated the EGF-induced keratinocyte migration, which was inhibited by the TRP channel blocker ruthenium red or by siRNA-mediated TRPV3 knockdown. Moreover, KS0365 induced strong Ca2+ responses in migrating front cells and in leading edges of keratinocytes. CONCLUSIONS AND IMPLICATIONS The selective TRPV3 activator KS0365 triggers increases in [Ca2+ ]i with most prominent signals in the leading edge, and accelerates migration of keratinocytes. TRPV3 activators may promote reepithelialization upon skin wounding.
Collapse
Affiliation(s)
- Marion Maier
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| | - Stefan Olthoff
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| | - Kerstin Hill
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| | - Carolin Zosel
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| | - Thomas Magauer
- Leopold-Franzens-University Innsbruck, Institute of Organic Chemistry and Center for Molecular Biosciences, Innsbruck, Austria
| | - Lukas Anton Wein
- Leopold-Franzens-University Innsbruck, Institute of Organic Chemistry and Center for Molecular Biosciences, Innsbruck, Austria
| | - Michael Schaefer
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| |
Collapse
|
15
|
Galunisertib Exerts Antifibrotic Effects on TGF-β-Induced Fibroproliferative Dermal Fibroblasts. Int J Mol Sci 2022; 23:ijms23126689. [PMID: 35743131 PMCID: PMC9223605 DOI: 10.3390/ijms23126689] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/10/2022] [Accepted: 06/11/2022] [Indexed: 02/06/2023] Open
Abstract
Dermal fibroblasts in pathological scars secrete constitutively elevated levels of TGF-β, signaling the transcription of fibrotic genes via activin-like kinase 5 (ALK5). In the present study, we examine the antifibrotic effects of galunisertib, a small-molecule inhibitor of ALK5, on fibroproliferative dermal fibroblasts in an in vitro model of wound healing. We induced fibrosis in human dermal fibroblasts with exogenous TGF-β and performed cellular proliferation assays after treatment with varying concentrations of galunisertib. Dermal fibroblast proliferation was diminished to homeostatic levels without cytotoxicity at concentrations as high as 10 μM. An in vitro scratch assay revealed that galunisertib significantly enhanced cellular migration and in vitro wound closure beginning 24 h post-injury. A gene expression analysis demonstrated a significant attenuation of fibrotic gene expression, including collagen-1a, alpha-smooth muscle actin, fibronectin, and connective tissue growth factor, with increased expression of the antifibrotic genes MMP1 and decorin. Protein synthesis assays confirmed drug activity and corroborated the transcription findings. In summary, galunisertib simultaneously exerts antifibrotic effects on dermal fibroblasts while enhancing rates of in vitro wound closure. Galunisertib has already completed phase II clinical trials for cancer therapy with minimal adverse effects and is a promising candidate for the treatment and prevention of pathological cutaneous scars.
Collapse
|
16
|
Eremenko E, Ding J, Kwan P, Tredget EE. The Biology of Extracellular Matrix Proteins in Hypertrophic Scarring. Adv Wound Care (New Rochelle) 2022; 11:234-254. [PMID: 33913776 DOI: 10.1089/wound.2020.1257] [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] [Indexed: 12/30/2022] Open
Abstract
Significance: Hypertrophic scars (HTS) are a fibroproliferative disorder that occur following deep dermal injury and affect up to 72% of burn patients. These scars result in discomfort, impaired mobility, disruption of normal function and cosmesis, and significant psychological distress. Currently, there are no satisfactory methods to treat or prevent HTS, as the cellular and molecular mechanisms are complex and incompletely understood. This review summarizes the biology of proteins in the dermal extracellular matrix (ECM), which are involved in wound healing and hypertrophic scarring. Recent Advances: New basic research continues toward understanding the diversity of cellular and molecular mechanisms of normal wound healing and hypertrophic scarring. Broadening the understanding of these mechanisms creates insight into novel methods for preventing and treating HTS. Critical Issues: Although there is an abundance of research conducted on collagen in the ECM and its relationship to HTS, there is a significant gap in understanding the role of proteoglycans and their specific isoforms in dermal fibrosis. Future Directions: Exploring the biological roles of ECM proteins and their unique isoforms in HTS, mature scars, and normal skin will further the understanding of abnormal wound healing and create a more robust understanding of what constitutes dermal fibrosis. Research into the biological roles of ECM protein isoforms and their regulation during wound healing warrants a more extensive investigation to identify their distinct biological functions in cellular processes and outcomes.
Collapse
Affiliation(s)
- Elizabeth Eremenko
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
| | - Jie Ding
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
| | - Peter Kwan
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
- Division of Plastic Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| | - Edward E. Tredget
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
- Division of Plastic Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| |
Collapse
|
17
|
Safina I, Childress LT, Myneni SR, Vang KB, Biris AS. Cell-Biomaterial Constructs for Wound Healing and Skin Regeneration. Drug Metab Rev 2022; 54:63-94. [PMID: 35129408 DOI: 10.1080/03602532.2021.2025387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Over the years, conventional skin grafts, such as full-thickness, split-thickness, and pre-sterilized grafts from human or animal sources, have been at the forefront of skin wound care. However, these conventional grafts are associated with major challenges, including supply shortage, rejection by the immune system, and disease transmission following transplantation. Due to recent progress in nanotechnology and material sciences, advanced artificial skin grafts-based on the fundamental concepts of tissue engineering-are quickly evolving for wound healing and regeneration applications, mainly because they can be uniquely tailored to meet the requirements of specific injuries. Despite tremendous progress in tissue engineering, many challenges and uncertainties still face skin grafts in vivo, such as how to effectively coordinate the interaction between engineered biomaterials and the immune system to prevent graft rejection. Furthermore, in-depth studies on skin regeneration at the molecular level are lacking; as a consequence, the development of novel biomaterial-based systems that interact with the skin at the core level has also been slow. This review will discuss 1) the biological aspects of wound healing and skin regeneration, 2) important characteristics and functions of biomaterials for skin regeneration applications, and 3) synthesis and applications of common biomaterials for skin regeneration. Finally, the current challenges and future directions of biomaterial-based skin regeneration will be addressed.
Collapse
Affiliation(s)
- Ingrid Safina
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR 72204 USA
| | - Luke T Childress
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR 72204 USA
| | - Srinivas R Myneni
- Department of Periodontology, Stony Brook University, Stony Brook, NY 11794 USA
| | - Kieng Bao Vang
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR 72204 USA
| | - Alexandru S Biris
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR 72204 USA
| |
Collapse
|
18
|
Fang L, Wu H, Li X, Fang J, Zhu Y. Improvement of Skin Wound Healing for Diabetic Mice with Thermosensitive Hydrogel Combined with Insulin Injection. Int J Endocrinol 2022; 2022:7847011. [PMID: 35311032 PMCID: PMC8930262 DOI: 10.1155/2022/7847011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/30/2022] [Accepted: 02/14/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic skin wound caused by diabetic disease is very common worldwide. Moreover, there is a shortage of effective curing technology in clinic. In this work, we developed a novel technology using thermosensitive hydrogel on wound top combined with insulin injection. The efficiency and mechanism of this technology were investigated in a diabetic mouse model. Dorsal-paired 8-10 mm diameter wounds were created in 12 mice. The wound healing rate was determined over a 28-day interval in healthy control (Control), control with diabetes (DControl), poloxamer treatment (Pox), and poloxamer plus insulin injection (Poxin) mice. Histological specimens were observed in all samples. Real-time quantitative polymerase chain reaction (qRT-PCR) was performed to measure the relative expression of α-smooth muscle actin (α-SMA) and transforming growth factor beta 1 (TGF-β1) in wound tissues at 7, 14, and 28 days. Compared with DControl animals, those treated with Poxin showed accelerated wound closure and healing rate (p < 0.05); expression of both α-SMA and TGF-β1 was significantly higher than that of the DControl and Pox animals during the first 7 days postoperation, but a significant decrease at day 14. Therefore, we concluded that hydrogel combined with insulin accelerated wound healing. Controlling the glucose level via insulin injection is more beneficial than hydrogel alone for healing chronic wounds, potentially through the increase of α-SMA and TGF-β1 expression in early phase.
Collapse
Affiliation(s)
- Lingling Fang
- Endocrinology Department, Ningbo Medical Center Lihuili Hospital, Affiliated to Ningbo University, 1111 Jiangnan Road, Ningbo 315000, China
| | - Haijian Wu
- School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo 315211, China
| | - Xiaoyan Li
- Medical Records and Statistics Office, Ningbo Medical Center Lihuili Hospital, 1111 Jiangnan Road, Ningbo 315000, China
| | - Jianghua Fang
- College of Material Science and Chemical Engineering, Ningbo University of Technology, 201 Fenghua Road, Ningbo 315211, China
| | - Yabin Zhu
- School of Medicine, Ningbo University, 818 Fenghua Road, Ningbo 315211, China
| |
Collapse
|
19
|
Zheng W, Lin G, Wang Z. Bioinformatics study on different gene expression profiles of fibroblasts and vascular endothelial cells in keloids. Medicine (Baltimore) 2021; 100:e27777. [PMID: 34964740 PMCID: PMC8615345 DOI: 10.1097/md.0000000000027777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 10/28/2021] [Indexed: 01/05/2023] Open
Abstract
Keloid is a benign fibroproliferative skin tumor. The respective functions of fibroblasts and vascular endothelial cells in keloid have not been fully studied. The purpose of this study is to identify the respective roles and key genes of fibroblasts and vascular endothelial cells in keloids, which can be used as new targets for diagnosis or treatment.The microarray datasets of keloid fibroblasts and vascular endothelial cells were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were screened out. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used for functional enrichment analysis. The search tool for retrieval of interacting genes and Cytoscape were used to construct protein-protein interaction (PPI) networks and analyze gene modules. The hub genes were screened out, and the relevant interaction networks and biological process analysis were carried out.In fibroblasts, the DEGs were significantly enriched in collagen fibril organization, extracellular matrix organization and ECM-receptor interaction. The PPI network was constructed, and the most significant module was selected, which is mainly enriched in ECM-receptor interaction. In vascular endothelial cells, the DEGs were significantly enriched in cytokine activity, growth factor activity and transforming growth factor-β (TGF-β) signaling pathway. Module analysis was mainly enriched in TGF-β signaling pathway. Hub genes were screened out separately.In summary, the DEGs and hub genes discovered in this study may help us understand the molecular mechanisms of keloid, and provide potential targets for diagnosis and treatment.
Collapse
Affiliation(s)
- Weihan Zheng
- School of Basic Medicine, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Guojian Lin
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, PR China
| | - Zhizhou Wang
- School of Basic Medicine, Fujian Medical University, Fuzhou, Fujian, PR China
| |
Collapse
|
20
|
Promotion of diabetic wound healing using novel Cu2O/Pt nanocubes through bacterial killing and enhanced angiogenesis in rats. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 134:112552. [DOI: 10.1016/j.msec.2021.112552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 11/21/2022]
|
21
|
Katayoshi T, Kusano Y, Shibata T, Uchida K, Tsuji-Naito K. Low-molecular-weight whey proteins promote collagen production in dermal fibroblasts via the TGF-β receptor/Smad pathway. Biosci Biotechnol Biochem 2021; 85:2232-2240. [PMID: 34498684 DOI: 10.1093/bbb/zbab155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/02/2021] [Indexed: 11/14/2022]
Abstract
Whey proteins (WPs) reportedly enhance cutaneous tissue regeneration in in vivo studies. However, the underlying mechanisms of such regenerative processes are poorly understood. In this study, we show that low-molecular-weight WPs (LMWPs; 1-30 kDa) accelerate the dermal collagen production via the transforming growth factor β receptor (TβR)/Smad pathway. We showed that LMWPs increased type I and III collagen expression in normal human dermal fibroblasts. Moreover, LMWPs rapidly induced Smad protein phosphorylation and nuclear translocation. Notably, type I TβR/Smad signaling inhibitor treatment or type II TβR siRNA knockdown blocked the LMWP-induced type I collagen expression. To identify the active components, we fractionated LMWPs and identified β-lactoglobulin and α-lactalbumin as potential TβR/Smad signaling inducers. Our findings unravel novel biological functions of WPs, involving the TβR/Smad-dependent induction of dermal collagen synthesis, highlighting the therapeutic potential of LMWPs in wound healing.
Collapse
Affiliation(s)
- Takeshi Katayoshi
- DHC Corporation Laboratories, Division 2, 2-42 Hamada, Mihama-ku, Chiba, Japan
| | - Yuri Kusano
- College of Bioscience and Biotechnology, Chubu University, 1200 Matsumoto-cho, Kasugai, Aichi, Japan
| | - Takahiro Shibata
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Koji Uchida
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kentaro Tsuji-Naito
- DHC Corporation Laboratories, Division 2, 2-42 Hamada, Mihama-ku, Chiba, Japan
| |
Collapse
|
22
|
Shanley LC, Mahon OR, Kelly DJ, Dunne A. Harnessing the innate and adaptive immune system for tissue repair and regeneration: Considering more than macrophages. Acta Biomater 2021; 133:208-221. [PMID: 33657453 DOI: 10.1016/j.actbio.2021.02.023] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/05/2021] [Accepted: 02/15/2021] [Indexed: 02/08/2023]
Abstract
Tissue healing and regeneration is a complex, choreographed, spatiotemporal process involving a plethora of cell types, the activity of which is stringently regulated in order for effective tissue repair to ensue post injury. A number of globally prevalent conditions such as heart disease, organ failure, and severe musculoskeletal disorders require new therapeutic strategies to repair damaged or diseased tissue, particularly given an ageing population in which obesity, diabetes, and consequent tissue defects have reached epidemic proportions. This is further compounded by the lack of intrinsic healing and poor regenerative capacity of certain adult tissues. While vast progress has been made in the last decade regarding tissue regenerative strategies to direct self-healing, for example, through implantation of tissue engineered scaffolds, several challenges have hampered the clinical application of these technologies. Control of the immune response is growing as an attractive approach in regenerative medicine and it is becoming increasingly apparent that an in depth understanding of the interplay between cells of the immune system and tissue specific progenitor cells is of paramount importance. Furthermore, the integration of immunology and bioengineering promises to elevate the efficacy of biomaterial-based tissue repair and regeneration. In this review, we highlight the role played by individual immune cell subsets in tissue repair processes and describe new approaches that are being taken to direct appropriate healing outcomes via biomaterial mediated targeting of immune cell activity. STATEMENT OF SIGNIFICANCE: It is becoming increasingly apparent that controlling the immune response is as an attractive approach in regenerative medicine. Here, we propose that an in-depth understanding of immune system and tissue specific progenitor cell interactions may reveal mechanisms by which tissue healing and regeneration takes place, in addition to identifying novel therapeutic targets that could be used to enhance the tissue repair process. To date, most reviews have focused solely on macrophage subsets. This manuscript details the role of other innate and adaptive immune cells such as innate lymphoid cells (ILCs), natural killer (NK) cells and γδT cells (in addition to macrophages) in tissue healing. We also describe new approaches that are being taken to direct appropriate healing outcomes via biomaterial mediated cytokine and drug delivery.
Collapse
|
23
|
Human Mesenchymal Stromal Cell-Derived Exosomes Promote In Vitro Wound Healing by Modulating the Biological Properties of Skin Keratinocytes and Fibroblasts and Stimulating Angiogenesis. Int J Mol Sci 2021; 22:ijms22126239. [PMID: 34207905 PMCID: PMC8228793 DOI: 10.3390/ijms22126239] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/31/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (MSCs) are major players in regenerative therapies for wound healing via their paracrine activity, mediated partially by exosomes. Our purpose was to test if MSC-derived exosomes could accelerate wound healing by enhancing the biological properties of the main cell types involved in the key phases of this process. Thus, the effects of exosomes on (i) macrophage activation, (ii) angiogenesis, (iii) keratinocytes and dermal fibroblasts proliferation and migration, and (iv) the capacity of myofibroblasts to regulate the turnover of the extracellular matrix were evaluated. The results showed that, although exosomes did not exhibit anti-inflammatory properties, they stimulated angiogenesis. Exposure of keratinocytes and dermal (myo)fibroblasts to exosomes enhanced their proliferation and migratory capacity. Additionally, exosomes prevented the upregulation of gene expression for type I and III collagen, α-smooth muscle actin, and MMP2 and 14, and they increased MMP13 expression during the fibroblast–myofibroblast transition. The regenerative properties of exosomes were validated using a wound healing skin organotypic model, which exhibited full re-epithelialization upon exosomes exposure. In summary, these data indicate that exosomes enhance the biological properties of keratinocytes, fibroblasts, and endothelial cells, thus providing a reliable therapeutic tool for skin regeneration.
Collapse
|
24
|
Mitra MS, Lancaster K, Adedeji AO, Palanisamy GS, Dave RA, Zhong F, Holdren MS, Turley SJ, Liang WC, Wu Y, Meng YG, Vernes JM, Schutten MM. A Potent Pan-TGFβ Neutralizing Monoclonal Antibody Elicits Cardiovascular Toxicity in Mice and Cynomolgus Monkeys. Toxicol Sci 2021; 175:24-34. [PMID: 32077954 DOI: 10.1093/toxsci/kfaa024] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Transforming growth factor β (TGFβ) signaling has been recently shown to reduce antitumor response to PD-L1 blockade, leading to a renewed enthusiasm in developing anti-TGFβ therapies for potential combination with cancer immunotherapy agents. Inhibition of TGFβ signaling in nonclinical toxicology species is associated with serious adverse toxicities including cardiac valvulopathies and anemia. Previously, cardiovascular toxicities have been thought to be limited to small molecule inhibitors of TGFβ receptor and not considered to be a liability associated with pan-TGFβ neutralizing monoclonal antibodies (mAbs). Here, we report the toxicity findings associated with a potent pan-TGFβ neutralizing mAb (pan-TGFβ mAb; neutralizes TGFβ1, 2, and 3) after 5 weekly intravenous doses of 10, 30, and 100 mg/kg, followed by a 4-week recovery period, in mice and cynomolgus monkeys. Mortality was observed due to acute bleeding and cardiovascular toxicity in mice at ≥ 30 mg/kg and prolonged menstruation in female monkeys at 100 mg/kg. Additional findings considered to be on-target exaggerated pharmacology included generalized bleeding and cardiovascular toxicity in mice and monkeys; histopathologic changes in the teeth, tongue, and skin in mice; and abnormal wound healing and microscopic pathology in the bone in monkeys. Importantly, our data indicate that the cardiovascular toxicities associated with the inhibition of TGFβ signaling are not limited to small molecule inhibitors but are also observed following administration of a potent pan-TGFβ inhibiting mAb.
Collapse
Affiliation(s)
- Mayur S Mitra
- Genentech Inc, South San Francisco, California 94080
| | | | | | | | - Rutwij A Dave
- Genentech Inc, South San Francisco, California 94080
| | - Fiona Zhong
- Genentech Inc, South San Francisco, California 94080
| | | | | | | | - Yan Wu
- Genentech Inc, South San Francisco, California 94080
| | - Y Gloria Meng
- Genentech Inc, South San Francisco, California 94080
| | | | | |
Collapse
|
25
|
Gori M, Vadalà G, Giannitelli SM, Denaro V, Di Pino G. Biomedical and Tissue Engineering Strategies to Control Foreign Body Reaction to Invasive Neural Electrodes. Front Bioeng Biotechnol 2021; 9:659033. [PMID: 34113605 PMCID: PMC8185207 DOI: 10.3389/fbioe.2021.659033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/27/2021] [Indexed: 12/21/2022] Open
Abstract
Neural-interfaced prostheses aim to restore sensorimotor limb functions in amputees. They rely on bidirectional neural interfaces, which represent the communication bridge between nervous system and neuroprosthetic device by controlling its movements and evoking sensory feedback. Compared to extraneural electrodes (i.e., epineural and perineural implants), intraneural electrodes, implanted within peripheral nerves, have higher selectivity and specificity of neural signal recording and nerve stimulation. However, being implanted in the nerve, their main limitation is represented by the significant inflammatory response that the body mounts around the probe, known as Foreign Body Reaction (FBR), which may hinder their rapid clinical translation. Furthermore, the mechanical mismatch between the consistency of the device and the surrounding neural tissue may contribute to exacerbate the inflammatory state. The FBR is a non-specific reaction of the host immune system to a foreign material. It is characterized by an early inflammatory phase eventually leading to the formation of a fibrotic capsule around intraneural interfaces, which increases the electrical impedance over time and reduces the chronic interface biocompatibility and functionality. Thus, the future in the reduction and control of the FBR relies on innovative biomedical strategies for the fabrication of next-generation neural interfaces, such as the development of more suitable designs of the device with smaller size, appropriate stiffness and novel conductive and biomimetic coatings for improving their long-term stability and performance. Here, we present and critically discuss the latest biomedical approaches from material chemistry and tissue engineering for controlling and mitigating the FBR in chronic neural implants.
Collapse
Affiliation(s)
- Manuele Gori
- Laboratory for Regenerative Orthopaedics, Department of Orthopaedic Surgery and Traumatology, Università Campus Bio-Medico di Roma, Rome, Italy
- Institute of Biochemistry and Cell Biology (IBBC) - National Research Council (CNR), Rome, Italy
| | - Gianluca Vadalà
- Laboratory for Regenerative Orthopaedics, Department of Orthopaedic Surgery and Traumatology, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Sara Maria Giannitelli
- Laboratory of Tissue Engineering, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Vincenzo Denaro
- Laboratory for Regenerative Orthopaedics, Department of Orthopaedic Surgery and Traumatology, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Giovanni Di Pino
- NeXT: Neurophysiology and Neuroengineering of Human-Technology Interaction Research Unit, Università Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
26
|
Chen L, Wu J, Hu B, Liu C, Wang H. The Role of Cell Division Autoantigen 1 (CDA1) in Renal Fibrosis of Diabetic Nephropathy. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6651075. [PMID: 33997036 PMCID: PMC8102118 DOI: 10.1155/2021/6651075] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 04/05/2021] [Accepted: 04/16/2021] [Indexed: 01/10/2023]
Abstract
The common kidney disease diabetic nephropathy (DN) accounts for significant morbidity and mortality in patients with diabetes, and its effective diagnosis in incipient stages is still lacking. Renal fibrosis is the main pathological feature of DN. Cell division autoantigen 1 (CDA1), a phosphorylated protein encoded by TSPYL2 on the X chromosome, plays a fibrogenic role by modulating the transforming growth factor-β (TGF-β) signaling, but the exact mechanism remains unclear. TGF-β signaling has been recognized as the key factor in promoting the development and progression of DN. At present, strict control of blood sugar and blood pressure can significantly lower the development and progression of DN in the early stages, and many studies have shown that blocking TGF-β signaling can delay the progress of DN. However, TGF-β is a multifunctional cytokine. Its direct intervention may result in increased side effects. Therefore, the targeted intervention of CDA1 not only can block the TGF-β signaling pathway but also can reduce these side effects. In this article, we review the main physiological roles of CDA1, with particular attention to its effect and potential mechanism in the renal fibrosis of DN.
Collapse
Affiliation(s)
- LinLin Chen
- Affiliated Ren He Hospital of China Three Gorges University, Yichang 443002, China
- Medical School, China Three Gorges University, 8 Daxue Road, Yichang 443002, China
| | - Jiao Wu
- Affiliated Ren He Hospital of China Three Gorges University, Yichang 443002, China
| | - Bin Hu
- Affiliated Ren He Hospital of China Three Gorges University, Yichang 443002, China
| | - Changbai Liu
- Medical School, China Three Gorges University, 8 Daxue Road, Yichang 443002, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, 8 Daxue Road, Yichang 443002, China
| | - Hu Wang
- Medical School, China Three Gorges University, 8 Daxue Road, Yichang 443002, China
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
27
|
Carnicer-Lombarte A, Chen ST, Malliaras GG, Barone DG. Foreign Body Reaction to Implanted Biomaterials and Its Impact in Nerve Neuroprosthetics. Front Bioeng Biotechnol 2021; 9:622524. [PMID: 33937212 PMCID: PMC8081831 DOI: 10.3389/fbioe.2021.622524] [Citation(s) in RCA: 179] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/19/2021] [Indexed: 12/04/2022] Open
Abstract
The implantation of any foreign material into the body leads to the development of an inflammatory and fibrotic process-the foreign body reaction (FBR). Upon implantation into a tissue, cells of the immune system become attracted to the foreign material and attempt to degrade it. If this degradation fails, fibroblasts envelop the material and form a physical barrier to isolate it from the rest of the body. Long-term implantation of medical devices faces a great challenge presented by FBR, as the cellular response disrupts the interface between implant and its target tissue. This is particularly true for nerve neuroprosthetic implants-devices implanted into nerves to address conditions such as sensory loss, muscle paralysis, chronic pain, and epilepsy. Nerve neuroprosthetics rely on tight interfacing between nerve tissue and electrodes to detect the tiny electrical signals carried by axons, and/or electrically stimulate small subsets of axons within a nerve. Moreover, as advances in microfabrication drive the field to increasingly miniaturized nerve implants, the need for a stable, intimate implant-tissue interface is likely to quickly become a limiting factor for the development of new neuroprosthetic implant technologies. Here, we provide an overview of the material-cell interactions leading to the development of FBR. We review current nerve neuroprosthetic technologies (cuff, penetrating, and regenerative interfaces) and how long-term function of these is limited by FBR. Finally, we discuss how material properties (such as stiffness and size), pharmacological therapies, or use of biodegradable materials may be exploited to minimize FBR to nerve neuroprosthetic implants and improve their long-term stability.
Collapse
Affiliation(s)
- Alejandro Carnicer-Lombarte
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Shao-Tuan Chen
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - George G. Malliaras
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Damiano G. Barone
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, United Kingdom
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
28
|
Ten Hove AS, Seppen J, de Jonge WJ. Neuronal innervation of the intestinal crypt. Am J Physiol Gastrointest Liver Physiol 2021; 320:G193-G205. [PMID: 33296267 DOI: 10.1152/ajpgi.00239.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mucosal damage is a key feature of inflammatory bowel diseases (IBD) and healing of the mucosa is an endpoint of IBD treatment that is often difficult to achieve. Autonomic neurons of the parasympathetic and sympathetic nervous system may influence intestinal epithelial cell growth and modulating epithelial innervation could for that reason serve as an interesting therapeutic option to improve mucosal healing. Understanding of the biological processes triggered by nonspecific and specific epithelial adrenergic and cholinergic receptor activation is of key importance. At present, with rising technological advances, bioelectronic neuromodulation as treatment modality has gained momentum. We discuss the current view on state-of-the-art innervation of the intestinal crypt and its impact on epithelial cell growth and differentiation. Furthermore, we outline bioelectronic technology and review its relevance to wound healing processes.
Collapse
Affiliation(s)
- Anne S Ten Hove
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Jurgen Seppen
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Wouter J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands.,Department of General, Visceral, Thoracic and Vascular Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
29
|
Janjetovic Z, Postlethwaite A, Kang HS, Kim TK, Tuckey RC, Crossman DK, Qayyum S, Jetten AM, Slominski AT. Antifibrogenic Activities of CYP11A1-derived Vitamin D3-hydroxyderivatives Are Dependent on RORγ. Endocrinology 2021; 162:bqaa198. [PMID: 33107570 PMCID: PMC7717072 DOI: 10.1210/endocr/bqaa198] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Indexed: 01/07/2023]
Abstract
Previous studies showed that noncalcemic 20(OH)D3, a product of CYP11A1 action on vitamin D3, has antifibrotic activity in human dermal fibroblasts and in a bleomycin mouse model of scleroderma. In this study, we tested the role of retinoic acid-related orphan receptor γ (RORγ), which is expressed in skin, in the action of CYP11A1-derived secosteroids using murine fibroblasts isolated from the skin of wild-type (RORγ +/+), knockout (RORγ -/-), and heterozygote (RORγ +/-) mice. CYP11A1-derived 20(OH)D3, 20,23(OH)2D3, 1,20(OH)2D3, and 1,20,23(OH)3D3 inhibited proliferation of RORγ +/+ fibroblasts in a dose-dependent manner with a similar potency to 1,25(OH)2D3. Surprisingly, this effect was reversed in RORγ +/- and RORγ -/- fibroblasts, with the most pronounced stimulatory effect seen in RORγ -/- fibroblasts. All analogs tested inhibited TGF-β1-induced collagen synthesis in RORγ +/+ fibroblasts and the expression of other fibrosis-related genes. This effect was curtailed or reversed in RORγ -/- fibroblasts. These results show that the antiproliferative and antifibrotic activities of the vitamin D hydroxy derivatives are dependent on a functional RORγ. The dramatic changes in the transcriptomes of fibroblasts of RORγ -/- versus wild-type mice following treatment with 20(OH)D3 or 1,20(OH)2D3 provide a molecular basis to explain, at least in part, the observed phenotypic differences.
Collapse
Affiliation(s)
- Zorica Janjetovic
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Arnold Postlethwaite
- Department of Medicine, University of Tennessee Health Science Center, Veteran Administration Medical Center, Memphis, Tennessee
| | - Hong Soon Kang
- Cell Biology Section, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Tae-Kang Kim
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Robert C Tuckey
- School of Molecular Sciences, the University of Western Australia, Crawley, Western Australia, Australia
| | | | - Shariq Qayyum
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anton M Jetten
- Cell Biology Section, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Andrzej T Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
- VA Medical Center, Birmingham, Alabama
| |
Collapse
|
30
|
Riley LA, Merryman WD. Cadherin-11 and cardiac fibrosis: A common target for a common pathology. Cell Signal 2020; 78:109876. [PMID: 33285242 DOI: 10.1016/j.cellsig.2020.109876] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023]
Abstract
Cardiac fibrosis represents an enormous health concern as it is prevalent in nearly every form of cardiovascular disease, the leading cause of death worldwide. Fibrosis is characterized by the activation of fibroblasts into myofibroblasts, a contractile cell type that secretes significant amounts of extracellular matrix components; however, the onset of this condition is also due to persistent inflammation and the cellular responses to a changing mechanical environment. In this review, we provide an overview of the pro-fibrotic, pro-inflammatory, and biomechanical mechanisms that lead to cardiac fibrosis in cardiovascular diseases. We then discuss cadherin-11, an intercellular adhesion protein present on both myofibroblasts and inflammatory cells, as a potential link for all three of the fibrotic mechanisms. Since experimentally blocking cadherin-11 dimerization prevents fibrotic diseases including cardiac fibrosis, understanding how this protein can be targeted for therapeutic use could lead to better treatments for patients with heart disease.
Collapse
Affiliation(s)
- Lance A Riley
- Department of Biomedical Engineering, Vanderbilt University, USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, USA.
| |
Collapse
|
31
|
|
32
|
Jackson JE, Kopecki Z, Anderson PJ, Cowin AJ. In vitro analysis of the effect of Flightless I on murine tenocyte cellular functions. J Orthop Surg Res 2020; 15:170. [PMID: 32398080 PMCID: PMC7216515 DOI: 10.1186/s13018-020-01692-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/29/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Healing of tendons after injury involves the proliferation of tenocytes and the production of extracellular matrix; however, their capacity to heal is limited by poor cell density and limited growth factor activity. Flightless I (Flii) has previously been identified as an important regulator of cellular proliferation and migration, and the purpose of this study was to evaluate the effect of differential Flii gene expression on tenocyte function in vitro. METHODS The role of Flii on tenocyte proliferation, migration, and contraction was assessed using established assays. Tenocytes from Flii+/-, wild-type, and Flii overexpressing mice were obtained and the effect of differential Flii expression on migration, proliferation, contraction, and collagen synthesis determined in vitro. Statistical differences were determined using unpaired Student's t test and statistical outliers were identified using the Grubbs' test. RESULTS Flii overexpressing tenocytes showed significantly improved migration and proliferation as well as increased collagen I secretion. Explanted tendons from Flii overexpressing mice also showed significantly elevated tenocyte outgrowth compared to Flii+/- mice. In contrast to its role in dermal wound repair, Flii positively affects cellular processes in tendons. CONCLUSIONS These findings suggest that Flii could be a novel target for modulating tenocyte activity and improving tendon repair. This could have significant clinical implications as novel therapeutic targets for improved healing of tendon injuries are urgently needed.
Collapse
Affiliation(s)
- Jessica E Jackson
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Zlatko Kopecki
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Peter J Anderson
- Faculty of Medicine and Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Allison J Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia.
| |
Collapse
|
33
|
Liu P, Choi JW, Lee MK, Choi YH, Nam TJ. Spirulina protein promotes skin wound repair in a mouse model of full-thickness dermal excisional wound. Int J Mol Med 2020; 46:351-359. [PMID: 32319537 PMCID: PMC7255466 DOI: 10.3892/ijmm.2020.4571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/02/2020] [Indexed: 11/11/2022] Open
Abstract
The skin protects body from environmental damage. Skin wounds lead to microbial infection and harmful agent injury. Thus, wound repair is crucial for the recovery of the normal function of skin tissue. The present study investigated the promoting effects of spirulina protein (SPCP) in mice on skin wound repair and also aimed to elucidate the potential underlying mechanisms. The results revealed that SPCP promoted the skin wound repair in a mouse model of full-thickness excisional wounds. SPCP induced an increase in the expression level of α-smooth muscle actin (α-SMA). The activities of superoxide dismutase (SOD) and catalase (CAT) were enhanced by SPCP treatment in the granulation tissue. In addition, SPCP decreased the level of malondialdehyde (MDA) in the granulation tissue. Western blot analysis revealed that SPCP enhanced the phosphorylation and activation of protein kinase B (Akt) and extracellular signal-regulated kinase (ERK). Moreover, the expression level of transforming growth factor β1 (TGF-β1) was increased in the SPCP-treated groups. The phosphorylation level of Smad2 was also increased by treatment of SPCP. Furthermore, SPCP promoted the expression of collagen in the granulation tissue. Taken together, these findings indicate that SPCP exerts a promoting effect on skin wound repair. The Akt, ERK and TGF-β1 signaling pathways are involved in this process.
Collapse
Affiliation(s)
- Ping Liu
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - Jeong-Wook Choi
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| | - Min-Kyeong Lee
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| | - Youn Hee Choi
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| | - Taek-Jeong Nam
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
34
|
Role of TGF-β in Skin Chronic Wounds: A Keratinocyte Perspective. Cells 2020; 9:cells9020306. [PMID: 32012802 PMCID: PMC7072438 DOI: 10.3390/cells9020306] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/17/2020] [Accepted: 01/27/2020] [Indexed: 12/19/2022] Open
Abstract
Chronic wounds are characterized for their incapacity to heal within an expected time frame. Potential mechanisms driving this impairment are poorly understood and current hypotheses point to the development of an unbalanced milieu of growth factor and cytokines. Among them, TGF-β is considered to promote the broadest spectrum of effects. Although it is known to contribute to healthy skin homeostasis, the highly context-dependent nature of TGF-β signaling restricts the understanding of its roles in healing and wound chronification. Historically, low TGF-β levels have been suggested as a pattern in chronic wounds. However, a revision of the available evidence in humans indicates that this could constitute a questionable argument. Thus, in chronic wounds, divergences regarding skin tissue compartments seem to be characterized by elevated TGF-β levels only in the epidermis. Understanding how this aspect affects keratinocyte activities and their capacity to re-epithelialize might offer an opportunity to gain comprehensive knowledge of the involvement of TGF-β in chronic wounds. In this review, we compile existing evidence on the roles played by TGF-β during skin wound healing, with special emphasis on keratinocyte responses. Current limitations and future perspectives of TGF-β research in chronic wounds are discussed.
Collapse
|
35
|
Wang Y, Graves DT. Keratinocyte Function in Normal and Diabetic Wounds and Modulation by FOXO1. J Diabetes Res 2020; 2020:3714704. [PMID: 33195703 PMCID: PMC7641706 DOI: 10.1155/2020/3714704] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 02/08/2023] Open
Abstract
Diabetes has a significant and negative impact on wound healing, which involves complex interactions between multiple cell types. Keratinocytes play a crucial role in the healing process by rapidly covering dermal and mucosal wound surfaces to reestablish an epithelial barrier with the outside environment. Keratinocytes produce multiple factors to promote reepithelialization and produce factors that enhance connective tissue repair through the elaboration of mediators that stimulate angiogenesis and production of connective tissue matrix. Among the factors that keratinocytes produce to aid healing are transforming growth factor-β (TGF-β), vascular endothelial growth factor-A (VEGF-A), connective tissue growth factor (CTGF), and antioxidants. In a diabetic environment, this program is disrupted, and keratinocytes fail to produce growth factors and instead switch to a program that is detrimental to healing. Changes in keratinocyte behavior have been linked to high glucose and advanced glycation end products that alter the activities of the transcription factor, FOXO1. This review examines reepithelialization and factors produced by keratinocytes that upregulate connective tissue healing and angiogenesis and how they are altered by diabetes.
Collapse
Affiliation(s)
- Yulan Wang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079 Hubei, China
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104 Pennsylvania, USA
- Department of Implantology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079 Hubei, China
| | - Dana T. Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104 Pennsylvania, USA
| |
Collapse
|
36
|
Losi P, Barsotti MC, Foffa I, Buscemi M, De Almeida CV, Fabbri M, Gabbriellini S, Nocchi F, Ursino S, Urciuoli P, Mazzoni A, Soldani G. In vitro human cord blood platelet lysate characterisation with potential application in wound healing. Int Wound J 2019; 17:65-72. [PMID: 31665826 DOI: 10.1111/iwj.13233] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/06/2019] [Accepted: 09/13/2019] [Indexed: 01/19/2023] Open
Abstract
Platelets contain abundant growth factors and cytokines that have a positive influence on the migration and proliferation of different cell types by modulating its physiopathological processes. As it is known that human umbilical cord blood platelet lysate (UCB-PL) contains a supraphysiological concentration of growth factors, in the present study, we investigated its effectiveness in wound-healing processes. Human UCB-PL was obtained by the freeze/thaw of platelet concentrate (1.1 × 109 platelets/L), and its effect was evaluated on human or mouse endothelial cells, monocytes, fibroblasts, and keratinocytes in different concentrations. Human UCB-PL was observed to have high levels of pro-angiogenic growth factor than peripheral blood platelet-rich plasma. Among the cell lines, different concentrations of human UCB-PL were necessary to influence their viability and proliferation. For L929 cells, 5% of total volume was necessary, while for human umbilical vein endothelial cell, it was 10%. Cell migration on monocytes was increased with respect to the positive control, and scratch closure on keratinocytes was increased with respect to serum-free medium with only 10% of human UCB-PL. We concluded that the human UCB-PL may be useful to produce a large amount of standard platelet concentrates sufficient for several clinical-scale expansions avoiding inter-individual variability, which can also be used as a functional tool for clinical regenerative application for wound healing.
Collapse
Affiliation(s)
- Paola Losi
- Laboratorio di Medicina Rigenerativa, Biomateriali e terapie avanzate, Institute of Clinical Physiology, National Research Council, Massa, Italy
| | - Maria C Barsotti
- Laboratorio di Medicina Rigenerativa, Biomateriali e terapie avanzate, Institute of Clinical Physiology, National Research Council, Massa, Italy
| | - Ilenia Foffa
- Laboratorio di Medicina Rigenerativa, Biomateriali e terapie avanzate, Institute of Clinical Physiology, National Research Council, Massa, Italy
| | - Marianna Buscemi
- Laboratorio di Medicina Rigenerativa, Biomateriali e terapie avanzate, Institute of Clinical Physiology, National Research Council, Massa, Italy
| | - Carolina V De Almeida
- Laboratorio di Medicina Rigenerativa, Biomateriali e terapie avanzate, Institute of Clinical Physiology, National Research Council, Massa, Italy
| | - Marco Fabbri
- Laboratorio di Immunogenetica, Medicina Trasfusionale e Biologia dei Trapianti, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Sabrina Gabbriellini
- Laboratorio di Immunogenetica, Medicina Trasfusionale e Biologia dei Trapianti, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Francesca Nocchi
- Laboratorio di Immunogenetica, Medicina Trasfusionale e Biologia dei Trapianti, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Silvia Ursino
- Laboratorio di Immunogenetica, Medicina Trasfusionale e Biologia dei Trapianti, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Patrizia Urciuoli
- Laboratorio di Immunogenetica, Medicina Trasfusionale e Biologia dei Trapianti, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Alessandro Mazzoni
- Medicina Trasfusionale e Biologia dei Trapianti, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Giorgio Soldani
- Laboratorio di Medicina Rigenerativa, Biomateriali e terapie avanzate, Institute of Clinical Physiology, National Research Council, Massa, Italy
| |
Collapse
|
37
|
Cakin MC, Ozdemir B, Kaya-Dagistanli F, Arkan H, Bahtiyar N, Anapali M, Akbas F, Onaran I. Evaluation of the in vivo wound healing potential of the lipid fraction from activated platelet-rich plasma. Platelets 2019; 31:513-520. [PMID: 31524041 DOI: 10.1080/09537104.2019.1663805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Previous in vitro studies suggest a direct relevance for the peptide-free lipid fraction (LF) of platelet-rich plasma (PRP) in biological mechanisms related to wound healing. However, there are no scientific reports to date on the wound healing activities of this lipid component in vivo. Thus, the present study provides a scientific evaluation for the wound healing potential of the lipid portion of the activated PRP. For the wound healing activity assessment, in vivo full-thickness excisional wounds were created on the dorsal skin of Sprague-Dawley rats. Lipid extract from pooled PRP was applied topically to the wounds on 0, 3, and 7 days after injury. Histological assessment of epidermal and dermal regeneration, granulation tissue thickness and angiogenesis by Sirius red and Masson's trichrome staining, in addition to immunohistochemical staining for transforming growth factor beta-1 (TGF-β1), collagen type I (COL I), and collagen type III (COL III) were performed on skin biopsies at 3, 7 and 14 days. The total histological scores of the LF group were significantly higher than the 25% dimethylsulfoxide-control group. According to the immunohistochemical staining, the observed expression changes for TGF-β1, COL I and III at 3, 7, and 14 days after wounding were significantly better in the study group than the control group. Furthermore, COL I/III ratio in the lipid extract-treated group at day 14 was much higher than that of the control group. Meanwhile, analysis of the data also indicated that the LF has less positive effects on all evaluated parameters than PRP. From the present data, it could be concluded that the peptide-free LF of PRP has potent wound healing capacity in vivo for cutaneous wounds, although not as much as that of PRP. Strengthening our understanding of the wound healing potential of lipid components of PRP and platelet-derived lipid factors may provide new avenues for improving the healing process of a wound with elevated protease activity.
Collapse
Affiliation(s)
- Mehmet Can Cakin
- Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa , Istanbul, Turkey
| | - Busra Ozdemir
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakif University , Istanbul, Turkey
| | - Fatma Kaya-Dagistanli
- Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa , Istanbul, Turkey
| | - Hulya Arkan
- Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa , Istanbul, Turkey
| | - Nurten Bahtiyar
- Department of Biophysics, Cerrahpasa Faculty of Medicine, Istanbul University- Cerrahpasa , Istanbul, Turkey
| | - Merve Anapali
- Department of Medical Biology, Faculty of Medicine, Ataturk University , Erzurum, Turkey
| | - Fahri Akbas
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakif University , Istanbul, Turkey
| | - Ilhan Onaran
- Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa , Istanbul, Turkey
| |
Collapse
|
38
|
Coufal S, Galanova N, Bajer L, Gajdarova Z, Schierova D, Jiraskova Zakostelska Z, Kostovcikova K, Jackova Z, Stehlikova Z, Drastich P, Tlaskalova-Hogenova H, Kverka M. Inflammatory Bowel Disease Types Differ in Markers of Inflammation, Gut Barrier and in Specific Anti-Bacterial Response. Cells 2019; 8:cells8070719. [PMID: 31337064 PMCID: PMC6678638 DOI: 10.3390/cells8070719] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/06/2019] [Accepted: 07/11/2019] [Indexed: 12/12/2022] Open
Abstract
Crohn’s disease (CD), ulcerative colitis (UC) and inflammatory bowel disease (IBD) associated with primary sclerosing cholangitis (PSC-IBD), share three major pathogenetic mechanisms of inflammatory bowel disease (IBD)-gut dysbiosis, gut barrier failure and immune system dysregulation. While clinical differences among them are well known, the underlying mechanisms are less explored. To gain an insight into the IBD pathogenesis and to find a specific biomarker pattern for each of them, we used protein array, ELISA and flow cytometry to analyze serum biomarkers and specific anti-microbial B and T cell responses to the gut commensals. We found that decrease in matrix metalloproteinase (MMP)-9 and increase in MMP-14 are the strongest factors discriminating IBD patients from healthy subjects and that PSC-IBD patients have higher levels of Mannan-binding lectin, tissue inhibitor of metalloproteinases 1 (TIMP-1), CD14 and osteoprotegerin than patients with UC. Moreover, we found that low transforming growth factor-β1 (TGF-β1) is associated with disease relapse and low osteoprotegerin with anti-tumor necrosis factor-alpha (TNF-α) therapy. Patients with CD have significantly decreased antibody and increased T cell response mainly to genera Eubacterium, Faecalibacterium and Bacteroides. These results stress the importance of the gut barrier function and immune response to commensal bacteria and point at the specific differences in pathogenesis of PSC-IBD, UC and CD.
Collapse
Affiliation(s)
- Stepan Coufal
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Natalie Galanova
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Lukas Bajer
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
- Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21 Prague, Czech Republic
| | - Zuzana Gajdarova
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Dagmar Schierova
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | | | - Klara Kostovcikova
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Zuzana Jackova
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Zuzana Stehlikova
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Pavel Drastich
- Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21 Prague, Czech Republic
| | - Helena Tlaskalova-Hogenova
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Miloslav Kverka
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic.
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic.
| |
Collapse
|
39
|
Lee MJ, Lee D, Jung HS. Wound healing mechanism in Mongolian gerbil skin. Histochem Cell Biol 2018; 151:229-238. [DOI: 10.1007/s00418-018-1752-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2018] [Indexed: 12/18/2022]
|
40
|
Park MK, Kim HJ, Cho MK, Kang SA, Park SY, Jang SB, Yu HS. Identification of a host collagen inducing factor from the excretory secretory proteins of Trichinella spiralis. PLoS Negl Trop Dis 2018; 12:e0006516. [PMID: 30383752 PMCID: PMC6233931 DOI: 10.1371/journal.pntd.0006516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 11/13/2018] [Accepted: 09/04/2018] [Indexed: 12/31/2022] Open
Abstract
Background In a previous study, we found that Trichinella spiralis muscle larva excretory and secretory proteins (ES-P) most likely activate collagen synthesis via TGF-β/Smad signaling, and this event could influence collagen capsule formation. Methodology/Principal findings In order to identify the specific collagen inducing factor, ES-P was fractionated by a Superdex 200 10/300 GL column. We obtained three large fractions, F1, F2, and F3, but only F3 had collagen gene inducing ability. After immunoscreening, 10 collagen inducing factor candidates were identified. Among them, TS 15–1 and TS 15–2 were identical to the putative trypsin of T. spiralis. The deduced TS 15–1 (M.W. = 72 kDa) had two conserved catalytic motifs, an N-terminal Tryp_SPc domain (TS 15-1n) and a C-terminal Tryp_SPc domain (TS 15-1c). To determine their collagen inducing ability, recombinant proteins (rTS 15-1n and rTS 15-1c) were produced using the pET-28a expression system. TS 15–1 is highly expressed during the muscle larval stage and has strong antigenicity. We determined that rTS 15-1c could elevate collagen I via activation of the TGF-β1 signaling pathway in vitro and in vivo. Conclusion/Significance In conclusion, we identified a host collagen inducing factor from T. spiralis ES-P using immunoscreening and demonstrated its molecular characteristics and functions. Trichinella spiralis can make collagen capsules in host muscle cells during its life cycle, which encapsulates muscle stage larvae. Many investigators have tried to reveal the complex mechanism behind this collagen capsule architecture, and it has been suggested that several serine proteases in excretory-secretory proteins of the parasite are potential collagen capsule inducing factors. In addition, collagen synthesis is activated through the TGF-β/Smad signaling pathway and these events are closely related with protease activated receptor 2 which was activated by various serine proteases. In this study, we isolated and characterized a collagen gene expression inducer from T. spiralis ES-P using immunoscreening and investigated the candidate protein for its usefulness as a wound healing therapeutic agent.
Collapse
Affiliation(s)
- Mi Kyung Park
- Department of Parasitology School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Hae-Jin Kim
- Department of Parasitology School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Min Kyoung Cho
- Department of Parasitology School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Shin Ae Kang
- Department of Parasitology School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - So Young Park
- Department of Parasitology School of Medicine, Pusan National University, Yangsan, Republic of Korea
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan, Republic of Korea
| | - Se Bok Jang
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan, Republic of Korea
| | - Hak Sun Yu
- Department of Parasitology School of Medicine, Pusan National University, Yangsan, Republic of Korea
- * E-mail:
| |
Collapse
|
41
|
Crane MJ, Xu Y, Henry WL, Gillis SP, Albina JE, Jamieson AM. Pulmonary influenza A virus infection leads to suppression of the innate immune response to dermal injury. PLoS Pathog 2018; 14:e1007212. [PMID: 30138446 PMCID: PMC6107272 DOI: 10.1371/journal.ppat.1007212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/12/2018] [Indexed: 12/31/2022] Open
Abstract
The innate immune system is responsible for many important functions in the body including responding to infection, clearing cancerous cells, healing wounds, and removing foreign substances. Although many of these functions happen simultaneously in life, most laboratory studies of the innate immune response focus on one activity. How the innate immune system responds to concurrent insults in different parts of the body is not well understood. This study explores the impact of a lung infection on the cutaneous wound healing process. We used two complimentary models of injury: the excisional tail wound and subcutaneous implantation of polyvinyl alcohol (PVA) sponges. These models allow for assessment of the rate of closure and measurement of cellular and cytokine responses during acute wound healing, respectively. When mice with these healing wounds were infected with influenza A virus (IAV) in the lung there was a delay in wound healing. The viral lung infection suppressed the innate immune response in a healing wound, including cellular infiltrate, chemokines, growth factors, and cytokines. However, there was not a global immune suppression as there was an increase in inflammation systemically in mice with both infection and healing wounds compared to mice with only wounds or IAV infection. In addition, the lung immune response was largely unaffected indicating that responding to a lung infection is prioritized over a healing wound. This study introduces the concept of immune triage, in that when faced with multiple insults the immune system prioritizes responses. This paradigm likely applies to many situations that involve the innate immune system, and understanding how the innate immune system handles multiple insults is essential to understanding how it can efficiently clear pathogens while responding to other inflammatory events. In a natural setting, the innate immune system is frequently faced with multiple insults, against which it must mount overlapping inflammatory responses. We are interested in how the innate immune system deals with multiple, simultaneously occurring inflammatory insults, and if the response to one will take priority. For example, the innate immune system is essential in mediating both the early control of pathogen replication in infected tissue and in the early stages of wound healing. Pulmonary infections occur frequently in injured patient populations; therefore, we set out to determine the impact of a respiratory infection on a healing wound. To examine this, mice with healing dermal wounds were infected with influenza A virus (IAV), a common cause of viral pneumonia. We found that the innate immune response to the lung infection took priority at the expense of the healing wound, in that the initial control of viral replication in the lung was not impacted, while the wound healing response was suppressed. Very little work has been done examining how the immune response can respond to overlapping inflammatory insults. Our work shows that not all immune responses are created equal, and that the cells of the innate immune system are preferentially routed towards fighting a lung infection rather than the healing dermal wound. This apparent prioritization of the innate immune response opens up a new direction of study. It is relevant to many fields where competing insults may alter the disease state.
Collapse
Affiliation(s)
- Meredith J. Crane
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States of America
| | - Yun Xu
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States of America
| | - William L. Henry
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States of America
| | - Sean P. Gillis
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, United States of America
| | - Jorge E. Albina
- Department of Surgery, Rhode Island Hospital and the Warren Alpert School of Medicine of Brown University, Providence, Rhode Island, United States of America
| | - Amanda M. Jamieson
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, United States of America
- * E-mail:
| |
Collapse
|
42
|
Albright V, Xu M, Palanisamy A, Cheng J, Stack M, Zhang B, Jayaraman A, Sukhishvili SA, Wang H. Micelle-Coated, Hierarchically Structured Nanofibers with Dual-Release Capability for Accelerated Wound Healing and Infection Control. Adv Healthc Mater 2018; 7:e1800132. [PMID: 29683273 PMCID: PMC6347427 DOI: 10.1002/adhm.201800132] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 03/03/2018] [Indexed: 12/18/2022]
Abstract
Tailoring nanofibrous matrices-a material with much promise for wound healing applications-to simultaneously mitigate bacterial colonization and stimulate wound closure of infected wounds is highly desirable. To that end, a dual-releasing, multiscale system of biodegradable electrospun nanofibers coated with biocompatible micellar nanocarriers is reported. For wound healing, transforming growth factor-β1 is incorporated into polycaprolactone/collagen (PCL/Coll) nanofibers via electrospinning and the myofibroblastic differentiation of human dermal fibroblasts is locally stimulated. To prevent infection, biocompatible nanocarriers of polypeptide-based block copolymer micelles are deposited onto the surfaces of PCL/Coll nanofibers using tannic acid as a binding partner. Micelle-modified fibrous scaffolds are favorable for wound healing, not only supporting the attachment and spreading of fibroblasts comparable to those on noncoated nanofibers, but also significantly enhancing fibroblast migration. Micellar coatings can be loaded with gentamicin or clindamycin and exhibit antibacterial activity as measured by Petrifilm and zone of inhibition assays as well as time-dependent reduction of cellular counts of Staphylococcus aureus cultures. Moreover, delivery time of antibiotic dosage is tunable through the application of a novel modular approach. Altogether, this system holds great promise as an infection-mitigating, cell-stimulating, biodegradable skin graft for wound management and tissue engineering.
Collapse
Affiliation(s)
- Victoria Albright
- Department of Materials Science and Engineering, Texas A&M University, 575 Ross Street, College Station, TX 77843, USA
| | - Meng Xu
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point on the Hudson, Hoboken, NJ 07030, USA
| | - Anbazhagan Palanisamy
- Department of Materials Science and Engineering, Texas A&M University, 575 Ross Street, College Station, TX 77843, USA
| | - Jun Cheng
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point on the Hudson, Hoboken, NJ 07030, USA
| | - Mary Stack
- Department of Biomedical Engineering, Stevens Institute of Technology, 1 Castle Point on the Hudson, Hoboken, NJ 07030, USA
| | - Beilu Zhang
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point on the Hudson, Hoboken, NJ 07030, USA
| | - Arul Jayaraman
- Department of Chemical Engineering, Department of Biomedical Engineering, Texas A&M University
| | - Svetlana A. Sukhishvili
- Department of Materials Science and Engineering, Texas A&M University, 575 Ross Street, College Station, TX 77843, USA,
| | - Hongjun Wang
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point on the Hudson, Hoboken, NJ 07030, USA,
| |
Collapse
|
43
|
Xie X, Percipalle P. Elevated transforming growth factor β signaling activation in β-actin-knockout mouse embryonic fibroblasts enhances myofibroblast features. J Cell Physiol 2018; 233:8884-8895. [PMID: 29851084 PMCID: PMC6220129 DOI: 10.1002/jcp.26808] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/30/2018] [Indexed: 01/08/2023]
Abstract
Signaling by the transforming growth factor‐β (TGF‐β) is an essential pathway regulating a variety of cellular events. TGF‐β is produced as a latent protein complex and is required to be activated before activating the receptor. The mechanical force at the cell surface is believed to be a mechanism for latent TGF‐β activation. Using β‐actin null mouse embryonic fibroblasts as a model, in which actin cytoskeleton and cell‐surface biophysical features are dramatically altered, we reveal increased TGF‐β1 activation and the upregulation of TGF‐β target genes. In β‐actin null cells, we show evidence that the enhanced TGF‐β signaling relies on the active utilization of latent TGF‐β1 in the cell culture medium. TGF‐β signaling activation contributes to the elevated reactive oxygen species production, which is likely mediated by the upregulation of Nox4. The previously observed myofibroblast phenotype of β‐actin null cells is inhibited by TGF‐β signaling inhibition, while the expression of actin cytoskeleton genes and angiogenic phenotype are not affected. Together, our study shows a scenario that the alteration of the actin cytoskeleton and the consequent changes in cellular biophysical features lead to changes in cell signaling process such as TGF‐β activation, which in turn contributes to the enhanced myofibroblast phenotype.
Collapse
Affiliation(s)
- Xin Xie
- Biology Program, Science Division, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
| | - Piergiorgio Percipalle
- Biology Program, Science Division, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
44
|
Tumor-fibroblast interactions stimulate tumor vascularization by enhancing cytokine-driven production of MMP9 by tumor cells. Oncotarget 2018; 8:35592-35608. [PMID: 28423685 PMCID: PMC5482601 DOI: 10.18632/oncotarget.16022] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 02/24/2017] [Indexed: 12/31/2022] Open
Abstract
Advance-stage breast carcinomas include significant amounts of fibroblasts and infiltrating immune cells which have been implicated in tumor growth, recurrence, and response to therapy. The present study investigated the contribution of fibroblasts to tumor growth using direct tumor-fibroblast co-cultures and tumor xenograft models. Our findings revealed that fibroblasts enhance breast carcinoma growth by promoting the tumor vasculature via the MMP9-dependent mechanism. In tumor-fibroblast co-cultures, fibroblasts increased expression of TGF-β, TNF, and IL-1β cytokines in tumor cells. These cytokines cooperatively induced expression of matrix metalloproteinase MMP9 in tumor cells. Knockdown of MMP9 by shRNA significantly reduced tumor vascularization induced by fibroblasts. Mechanistically, our findings argue that expression of MMP9 in tumor cellsis regulated by crosstalk of TGF-β with TNF and/or IL-1β cytokines. The mechanism of this cooperative response did not involve cross-activation of the canonical signaling pathways as TGF-β did not activate RELA/p65 signaling, while TNF did not affect SMAD signaling. Instead, TGF-β and TNF cytokines co-stimulated MAP kinases and expression of JUN and JUNB, AP1 transcription factor subunits, which together with RELA/p65 were essential for the regulation of MMP9. Depletion of JUN and JUNB or RELA in tumor cells blocked the cooperative induction of MMP9 by the cytokines. Thus, our studies uncovered a previously unappreciated role of tumor-fibroblast interactions in the stimulation of tumor angiogenesis, and an essential role of the MAPK-AP1 axis in the cooperative up-regulation of the angiogenic driver MMP9 by cytokine crosstalk.
Collapse
|
45
|
Gao SQ, Chang C, Niu XQ, Li LJ, Zhang Y, Gao JQ. Topical application of Hydroxysafflor Yellow A accelerates the wound healing in streptozotocin induced T1DM rats. Eur J Pharmacol 2018; 823:72-78. [PMID: 29408092 DOI: 10.1016/j.ejphar.2018.01.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/12/2018] [Accepted: 01/15/2018] [Indexed: 12/13/2022]
Abstract
To investigate the effects of Hydroxysafflor Yellow A (HSYA), which is derived from safflower, on the proliferation, migration and angiogenesis of cells in vitro and its potential efficacy in vivo when topically applied to a diabetic wound. Human umbilical vein endothelial cells (HUVECs) and mouse macrophage cells (RAW264.7) were used to evaluate angiogenesis and anti-inflammatory activities, respectively. The influence of HSYA on the wound scratch assay was investigated in keratinocytes. A splinted excisional wound model in rats with TIDM induced by streptozotocin was used to assess the effects of wound healing. Collagen disposition and secretion of vascular growth factors (VEGF) as well as transforming growth factor-β1 (TGF-β1) were evaluated by an ELISA assay and histological staining. The in vitro results showed that HSYA could significantly enhance both the neovascularization of HUVECs and the migration of keratinocytes. It showed the significant inhibitory effect on nitric oxide production, indicating the anti-inflammatory activity of HSYA. In vivo, the topical application of HSYA significantly enhanced the wound closure rate, and the time to complete wound closure was 17 days, whereas 30 days were needed with PBS treatment. Further, treatment with HSYA exhibited significant granulation tissue formation with higher collagen content, re-epithelialization and angiogenesis according to Masson's trichrome staining evaluation, VEGE and TGF-β1 ELISA measurement. In conclusion, HSYA application could be considered a promising therapeutic strategy for treating chronic non-healing diabetic foot ulcers.
Collapse
Affiliation(s)
- Si-Qian Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, PR China
| | - Chen Chang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, PR China
| | - Xiao-Qian Niu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, PR China
| | - Long-Jian Li
- Zhejiang Provincial Corps Hospital of Chinese People's Armed Police Forces, Jiaxing, Zhejiang, PR China
| | - Yan Zhang
- Zhejiang Provincial Corps Hospital of Chinese People's Armed Police Forces, Jiaxing, Zhejiang, PR China
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, PR China; Jiangsu Engineering Research Center for New-Type External and Transdermal Preparations, PR China.
| |
Collapse
|
46
|
Abadir P, Hosseini S, Faghih M, Ansari A, Lay F, Smith B, Beselman A, Vuong D, Berger A, Tian J, Rini D, Keenahan K, Budman J, Inagami T, Fedarko N, Marti G, Harmon J, Walston J. Topical Reformulation of Valsartan for Treatment of Chronic Diabetic Wounds. J Invest Dermatol 2018; 138:434-443. [PMID: 29078982 PMCID: PMC10941026 DOI: 10.1016/j.jid.2017.09.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 09/05/2017] [Accepted: 09/07/2017] [Indexed: 02/07/2023]
Abstract
Chronic wounds are among the most devastating and difficult to treat consequences of diabetes. Dysregulation of the skin renin-angiotensin system is implicated in abnormal wound healing in diabetic and older adults. Given this, we sought to determine the effects of topical reformulations of the angiotensin type 1 receptor blockers losartan and valsartan and the angiotensin-converting enzyme inhibitor captopril on wound healing in diabetic and aged mice with further validation in older diabetic pigs. The application of 1% valsartan gel compared with other tested formulations and placebo facilitated and significantly accelerated closure time and increased tensile strength in mice, and was validated in the porcine model. One percent of valsartan gel-treated wounds also exhibited higher mitochondrial content, collagen deposition, phosphorylated mothers against decapentaplegic homologs 2 and 3 and common mothers against decapentaplegic homolog 4, alpha-smooth muscle actin, CD31, phospho-vascular endothelial growth factor receptor 2, and p42/44 mitogen-activated protein kinase. Knockout of the angiotensin subtype 2 receptors abolished the beneficial effects of angiotensin type 1 receptor blockers, suggesting a role for angiotensin subtype 2 receptors in chronic wound healing.
Collapse
Affiliation(s)
- Peter Abadir
- Division of Geriatrics Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Sayed Hosseini
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mahya Faghih
- Division of Geriatrics Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amir Ansari
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Frank Lay
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Barbara Smith
- Cell Biology Imaging Facility, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aleksandra Beselman
- Investigational Drug Service Pharmacy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Diep Vuong
- Division of Geriatrics Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alan Berger
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jing Tian
- Department of Biostatistics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David Rini
- Art as Applied to Medicine, Division of Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kevin Keenahan
- Department of Bioengineering Innovation, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joshua Budman
- Department of Bioengineering Innovation, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tadashi Inagami
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Neal Fedarko
- Division of Geriatrics Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guy Marti
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Clinique Saint Jean, Melun, France
| | - John Harmon
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeremy Walston
- Division of Geriatrics Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
47
|
Amini-Nik S, Yousuf Y, Jeschke MG. Scar management in burn injuries using drug delivery and molecular signaling: Current treatments and future directions. Adv Drug Deliv Rev 2018; 123:135-154. [PMID: 28757325 PMCID: PMC5742037 DOI: 10.1016/j.addr.2017.07.017] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/14/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022]
Abstract
In recent decades, there have been tremendous improvements in burn care that have allowed patients to survive severe burn injuries that were once fatal. However, a major limitation of burn care currently is the development of hypertrophic scars in approximately 70% of patients. This significantly decreases the quality of life for patients due to the physical and psychosocial symptoms associated with scarring. Current approaches to manage scarring include surgical techniques and non-surgical methods such as laser therapy, steroid injections, and compression therapy. These treatments are limited in their effectiveness and regularly fail to manage symptoms. As a result, the development of novel treatments that aim to improve outcomes and quality of life is imperative. Drug delivery that targets the molecular cascades of wound healing to attenuate or prevent hypertrophic scarring is a promising approach that has therapeutic potential. In this review, we discuss current treatments for scar management after burn injury, and how drug delivery targeting molecular signaling can lead to new therapeutic strategies.
Collapse
Affiliation(s)
- Saeid Amini-Nik
- Sunnybrook Research Institute, Toronto, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada.
| | - Yusef Yousuf
- Institute of Medical Science, University of Toronto, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada
| | - Marc G Jeschke
- Institute of Medical Science, University of Toronto, Toronto, Canada; Sunnybrook Research Institute, Toronto, Canada; Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada; Department of Immunology, University of Toronto, Toronto, Canada; Ross-Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Canada.
| |
Collapse
|
48
|
Nunomura S, Nanri Y, Ogawa M, Arima K, Mitamura Y, Yoshihara T, Hasuwa H, Conway SJ, Izuhara K. Constitutive overexpression of periostin delays wound healing in mouse skin. Wound Repair Regen 2018; 26:6-15. [PMID: 29418037 PMCID: PMC5906136 DOI: 10.1111/wrr.12616] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/22/2018] [Indexed: 12/28/2022]
Abstract
Periostin is a matricellular protein involved in development, maintenance, and regulation of tissues and organs via by binding to cell surface integrin receptors. Pathologically, periostin plays an important role in the process of wound healing: as a deficiency of the Postn gene delays wound closure and periostin is consistently up-regulated in response to injury and skin diseases. However, the functional role of elevated periostin in the process of wound healing has not been tested. In this study, we generated Postn-transgenic mice under the control of the CAG promoter/enhancer to investigate the effects of constitutive overexpression of full length periostin during its pathophysiological roles. Transgenic mice showed significant overexpression of periostin in skin, lung, and heart, but no morphological changes were observed. However, when these transgenic mice were injured, periostin overexpression delayed the closure of excisional wounds. Expression of IL-1β and TNFα, pro-inflammatory cytokines important for wound healing, was significantly decreased in the transgenic mice, prior to delayed healing. Infiltration of neutrophils and macrophages, the main sources of IL-1β and TNFα, was also down-regulated in the transgenic wound sites. From these data, we conclude that enforced expression of periostin delays wound closure due to reduced infiltration of neutrophils and macrophages followed by down-regulation of IL-1β and TNFα expression. This suggests that regulated spatiotemporal expression of periostin is important for efficient wound healing and that constitutive periostin overexpression interrupts the normal process of wound closure.
Collapse
Affiliation(s)
- Satoshi Nunomura
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Yasuhiro Nanri
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Masahiro Ogawa
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Kazuhiko Arima
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Yasutaka Mitamura
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Tomohito Yoshihara
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Hidetoshi Hasuwa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Simon J. Conway
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kenji Izuhara
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, 5-1-1 Nabeshima, Saga 849-8501, Japan
| |
Collapse
|
49
|
Zheng Z, James AW, Li C, Jiang W, Wang JZ, Chang GX, Lee KS, Chen F, Berthiaume EA, Chen Y, Pan HC, Chen EC, Li W, Zhao Z, Zhang X, Ting K, Soo C. Fibromodulin reduces scar formation in adult cutaneous wounds by eliciting a fetal-like phenotype. Signal Transduct Target Ther 2017; 2:17050-. [PMID: 29201497 PMCID: PMC5661627 DOI: 10.1038/sigtrans.2017.50] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 05/28/2017] [Accepted: 08/18/2017] [Indexed: 02/02/2023] Open
Abstract
Blocking transforming growth factor (TGF)β1 signal transduction has been a central strategy for scar reduction; however, this approach appears to be minimally effective. Here, we show that fibromodulin (FMOD), a 59-kD small leucine-rich proteoglycan critical for normal collagen fibrillogenesis, significantly reduces scar formation while simultaneously increasing scar strength in both adult rodent models and porcine wounds, which simulate human cutaneous scar repair. Mechanistically, FMOD uncouples pro-migration/contraction cellular signals from pro-fibrotic signaling by selectively enhancing SMAD3-mediated signal transduction, while reducing AP-1-mediated TGFβ1 auto-induction and fibrotic extracellular matrix accumulation. Consequently, FMOD accelerates TGFβ1-responsive adult fibroblast migration, myofibroblast conversion, and function. Furthermore, our findings strongly indicate that, by delicately orchestrating TGFβ1 activities rather than indiscriminately blocking TGFβ1, FMOD elicits fetal-like cellular and molecular phenotypes in adult dermal fibroblasts in vitro and adult cutaneous wounds in vivo, which is a unique response of living system undescribed previously. Taken together, this study illuminates the signal modulating activities of FMOD beyond its structural support functions, and highlights the potential for FMOD-based therapies to be used in cutaneous wound repair.
Collapse
Affiliation(s)
- Zhong Zheng
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, The Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aaron W James
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, The Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Chenshuang Li
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Wenlu Jiang
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Joyce Z Wang
- Department of Emergency Medicine, Highland General Hospital, Oakland, CA 94602, USA
| | - Grace X Chang
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kevin S Lee
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Feng Chen
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Central Laboratory, School of Stomatology, Peking University, Beijing 100081, China
| | - Emily A Berthiaume
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yao Chen
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hsin Chuan Pan
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Eric C Chen
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Weiming Li
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Orthopaedics, the First Clinical Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xinli Zhang
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kang Ting
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, The Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chia Soo
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, The Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
50
|
Biochemical and functional analysis of corticotropin releasing factor purified from an aqueous extract of human placenta used as wound healer. J Pharm Biomed Anal 2017; 145:298-306. [PMID: 28704719 DOI: 10.1016/j.jpba.2017.06.060] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/23/2017] [Accepted: 06/27/2017] [Indexed: 12/18/2022]
Abstract
Human placental extract constitutes of innumerable therapeutically important components mostly used in wound healing arising from the skin and burn injuries. However, there is still some bioactive present in the placental extracts yet to be characterized to better under the complex process of wound healing mediated by the placental extract. In this study, the presence of corticotropin releasing factor (CRF) in an aqueous extract of human placenta was detected and quantified by dot blot and CRF-ELISA immunoassay kit respectively. Subsequently, it was purified by immuno-affinity chromatography and quantified as 0.45±0.05μg of CRF per ml of placental extract where its molecular weight found to be 4.78kDa by MALDI-TOF. To study functional analysis of CRF, an in vitro WI-38 lung fibroblast cell scratch wound model was used which indicated proliferation, motility of cells after treatment with purified CRF. Moreover, reduction in apoptosis rate of cells during closure of wound was observed from microscopy studies and FACS analysis. Also, Antalarmin, an antagonist of CRF type 1 receptor inhibited the wound closure potency of the purified component. Faster healing of wound with an elevation of IL-6 and TGF-β during early stages of repair by placental CRF was observed on excision rat model. The process of healing was accompanied by the decrease in the level of TNF-α and IFN-γ.
Collapse
|