1
|
Staianov J, Struz JMR, Vieira RV, Luiz RM, Zarpelon-Schutz AC, Teixeira KN, Bernardi-Wenzel J. Histomorphometric analysis of excisional cutaneous wounds with different diameters in an animal model. Int J Exp Pathol 2024. [PMID: 39439085 DOI: 10.1111/iep.12520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
The skin wound model in rats is a fundamental stage in preclinical trials, but there is a lack of standardization in these trials regarding the initial wound area, making analysis and comparison between studies difficult. Therefore, this study evaluates the healing progression of excisional skin lesions of varying diameters in Wistar rats, aiming to identify the optimal wound size for monitoring treatment effects on wound healing. Excisions of 0.8, 1.5, 2.0 and 3.0 cm in diameter were made on the back of the animals. Thirty animals were used per treatment and evaluated on days 3, 7, 10, 14 and 21 after surgery. The lesions were cleaned daily with saline solution until they were completely closed. The 0.8 cm group showed complete repair on D14, while in the other groups, the wounds persisted until day 21, with a reddened surface and no complete epidermal coverage, but with greater keratinization and presence of appendages in the 1.5 cm lesions. Therefore, as a standardization model for creating skin wounds, we suggest using 1.5 or 2.0 cm excisions, considering that 0.8 cm wounds close very early and 3.0 cm wounds, although behaving similarly to 2.0 cm wounds, are more invasive for the animals. The 1.5 cm model proved to be suitable for closure within 21 days. When evaluating a product intended to accelerate wound healing, 2.0 cm lesions are recommended to assess the effectiveness of the treatment.
Collapse
Affiliation(s)
- Janiele Staianov
- Curso de Medicina, Campus Toledo, Universidade Federal do Paraná, Toledo, Brazil
| | | | - Rafaela Viana Vieira
- Curso de Medicina, Campus Toledo, Universidade Federal do Paraná, Toledo, Brazil
| | - Rafael Messias Luiz
- Curso de Medicina, Campus Toledo, Universidade Federal do Paraná, Toledo, Brazil
| | - Ana Carla Zarpelon-Schutz
- Curso de Medicina, Campus Toledo, Universidade Federal do Paraná, Toledo, Brazil
- Programa de Pós-graduação em Biotecnologia, Setor Palotina, Universidade Federal do Paraná, Palotina, Brazil
| | - Kádima Nayara Teixeira
- Curso de Medicina, Campus Toledo, Universidade Federal do Paraná, Toledo, Brazil
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular, Setor Palotina, Universidade Federal do Paraná, Palotina, Brazil
| | - Juliana Bernardi-Wenzel
- Curso de Medicina, Campus Toledo, Universidade Federal do Paraná, Toledo, Brazil
- Programa de Pós-graduação em Biotecnologia, Setor Palotina, Universidade Federal do Paraná, Palotina, Brazil
| |
Collapse
|
2
|
Zhang Y, Fu R, Zhu C, Yuwen W, Zhang J, Duan Z, Fan D. Preparation of recombinant type I collagen (PF-I-80) and its functional characterization and biomedical applications in wound healing. Int J Biol Macromol 2024; 282:136679. [PMID: 39437939 DOI: 10.1016/j.ijbiomac.2024.136679] [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: 04/22/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
This study evaluates the potential applications of recombinant PF-I-80 protein in regenerative medicine and the treatment of inflammatory diseases, focusing on its effects on cell migration, differentiation, and anti-inflammatory properties. Various in vitro assays were conducted, including scratch assays, Transwell experiments, RT-PCR and Western Blot to analyze gene and protein expression related to differentiation and inflammation, and immunofluorescence staining to observe cellular changes. The results indicated that PF-I-80 significantly promoted cell migration, highlighting its potential in tissue repair and regeneration. It also enhanced cell differentiation, demonstrating its applicability in tissue repair, and showed significant anti-inflammatory effects by reducing the expression of pro-inflammatory cytokines. In animal models, PF-I-80 notably reduced levels of inflammatory factors IL-1β and TNF-α, shortened the inflammatory phase, and accelerated wound healing. Additionally, PF-I-80 increased FGF-2 levels, which promoted the proliferation of endothelial and fibroblast cells and enhanced collagen synthesis. These in vitro and in vivo findings position PF-I-80 as a promising biomaterial for applications in regenerative medicine and inflammatory disease treatment.
Collapse
Affiliation(s)
- Yan Zhang
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Rongzhan Fu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Chenhui Zhu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Weigang Yuwen
- Taibai Campus, Northwest University, Xi'an 710069, China
| | - Jiangrui Zhang
- Taibai Campus, Northwest University, Xi'an 710069, China
| | - Zhiguang Duan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China.
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China.
| |
Collapse
|
3
|
Ma H, Wang T, Li G, Liang J, Zhang J, Liu Y, Zhong W, Li P. A photo-modulated nitric oxide delivering hydrogel for the accelerated healing of biofilm infected chronic wounds. Acta Biomater 2024; 188:169-183. [PMID: 39299622 DOI: 10.1016/j.actbio.2024.09.017] [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: 03/21/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Biofilm infection and impaired healing of chronic wounds are posing tremendous challenges in clinical practice. In this study, we presented a versatile antimicrobial hydrogel capable of delivering nitric oxide (NO) in a controllable manner to dissipate biofilms, eliminate microorganisms, and promote the healing of chronic wounds. This hydrogel was constructed by Schiff-base crosslinking of oxidized dextran and antimicrobial peptide ε-poly-lysine, further encapsulating photothermal nanoparticles bearing NO donor. This hydrogel could continuously and slowly release NO, effectively dissipating biofilms, and promoting the proliferation of mouse fibroblasts and the migration of endothelial cells. Upon exposure to NIR laser irradiation, the hydrogel generated hyperthermia and rapidly released NO, resulting in the efficient elimination of a broad spectrum of drug-resistant Gram-positive/negative bacterial and fungal biofilms through the synergistic effects of NO, photothermal therapy, and the antibacterial peptide. Notably, the hydrogel demonstrated exceptional in vivo therapeutic outcomes in accelerating the healing process of mice diabetic wounds infected with methicillin-resistant Staphylococcus aureus by successfully eliminating biofilm infection, regulating inflammation, and facilitating angiogenesis and collagen deposition. Overall, this proposed hydrogel shows great promise in accommodating the various demands of the complex repair process of chronic wounds infected with biofilms. STATEMENT OF SIGNIFICANCE: The presence of biofilm infections and underlying dysfunctions in the healing process made chronic wound become stuck in the inflammation stage and difficult to heal. This work developed a NIR laser-modulated three-stage NO-releasing versatile antimicrobial hydrogel (DEPN) exhibiting good therapeutic efficacy for chronic wound. This DEPN hydrogel could inherently and slowly released NO to disperse biofilm. Upon NIR laser irradiation, the DEPN hydrogel generated hyperthermia and induced a rapid burst release of NO effectively eliminating a broad spectrum of drug-resistant bacterial and fungal biofilms. Subsequently, the DEPN hydrogel continually release NO slowly to promote the tissue remolding. This DEPN hydrogel displays great potential in treatment of chronic wounds infected with biofilm.
Collapse
Affiliation(s)
- Huifang Ma
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute, Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an 710072, China; School of Flexible Electronics (SoFE) and Henan Institute of Flexible Electronics (HIFE), Henan University, 379 Mingli Road, Zhengzhou 450046, China
| | - Tengjiao Wang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute, Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an 710072, China; School of Flexible Electronics (SoFE) and Henan Institute of Flexible Electronics (HIFE), Henan University, 379 Mingli Road, Zhengzhou 450046, China; Chongqing Technology Innovation Center, Northwestern Polytechnical University (NPU), Chongqing 401120, China.
| | - Gangfeng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute, Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an 710072, China
| | - Jiaheng Liang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute, Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an 710072, China
| | - Jianhong Zhang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute, Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an 710072, China
| | - Yang Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637371, Singapore
| | - Wenbin Zhong
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637371, Singapore
| | - Peng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute, Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an 710072, China; School of Flexible Electronics (SoFE) and Henan Institute of Flexible Electronics (HIFE), Henan University, 379 Mingli Road, Zhengzhou 450046, China.
| |
Collapse
|
4
|
Kosaka K, Takayama N, Paul SK, Kanashiro MA, Oshima M, Fukuyo M, Rahmutulla B, Tajiri I, Mukai M, Kubota Y, Akita S, Furuyama N, Kaneda A, Iwama A, Eto K, Mitsukawa N. iPSC-derived megakaryocytes and platelets accelerate wound healing and angiogenesis. Stem Cell Res Ther 2024; 15:364. [PMID: 39402677 PMCID: PMC11477011 DOI: 10.1186/s13287-024-03966-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Platelet-rich plasma (PRP), which is prepared by concentrating platelets in autologous blood, shows efficacy in chronic skin wounds via multiple growth factors. However, it exhibits heterogeneity across patients, leading to unstable therapeutic efficacy. Human induced pluripotent stem cell (iPSC)-derived megakaryocytes and platelets (iMPs) are capable of providing a stable supply, holding promise as materials for novel platelet concentrate-based therapies. In this context, we evaluated the effect of iMPs on wound healing and validated lyophilization for clinical applications. METHODS The growth factors released by activated iMPs were measured. The effect of the administration of iMPs on human fibroblasts and human umbilical vein endothelial cells (HUVECs) was investigated in vitro. iMPs were applied to dorsal skin defects of diabetic mice to assess the wound closure rate and quantify collagen deposition and angiogenesis. Following the storage of freeze-dried iMPs (FD-iMPs) for three months, the stability of growth factors and their efficacy in animal models were determined. RESULT Multiple growth factors that promote wound healing were detected in activated iMPs. iMPs specifically released FGF2 and exhibited a superior enhancement of HUVEC proliferation compared to PRP. Moreover, an RNA-seq analysis revealed that iMPs induce polarization to stalk cells and enhance ANGPTL4 gene expression in HUVECs. Animal studies demonstrated that iMPs promoted wound closure and angiogenesis in chronic wounds caused by diabetes. We also confirmed the long-term stability of growth factors in FD-iMPs and their comparable effects to those of original iMPs in the animal model. CONCLUSION Our study demonstrates that iMPs promote angiogenesis and wound healing through the activation of vascular endothelial cells. iMPs exhibited more effectiveness than PRP, an effect attributed to the exclusive presence of specific factors including FGF2. Lyophilization enabled the long-term maintenance of the composition of the growth factors and efficacy of the iMPs, therefore contributing to stable supply for clinical application. These findings suggest that iMPs provide a novel treatment for chronic wounds.
Collapse
Affiliation(s)
- Kentaro Kosaka
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan.
| | - Naoya Takayama
- Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Sudip Kumar Paul
- Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | - Motohiko Oshima
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masaki Fukuyo
- Department of Molecular Oncology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Bahityar Rahmutulla
- Department of Molecular Oncology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Ikuko Tajiri
- Department of Orthopedic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Michiaki Mukai
- Department of Orthopedic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yoshitaka Kubota
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shinsuke Akita
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | - Atsushi Kaneda
- Department of Molecular Oncology, Chiba University Graduate School of Medicine, Chiba, Japan
- Health and Disease Omics Center, Chiba University, Chiba, Japan
| | - Atsushi Iwama
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koji Eto
- Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, Japan.
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.
| | - Nobuyuki Mitsukawa
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan.
| |
Collapse
|
5
|
Toma AI, Shah D, Roth D, Piña JO, Hymel L, Turner T, Kamalakar A, Liu K, Bartsch P, Jacobs L, D'Souza R, Liotta D, Botchwey E, Willett NJ, Goudy SL. Accelerating Oral Wound Healing Using Bilayer Biomaterial Delivery of FTY720 Immunotherapy. Adv Healthc Mater 2024:e2401480. [PMID: 39388502 DOI: 10.1002/adhm.202401480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/27/2024] [Indexed: 10/12/2024]
Abstract
Orofacial clefts are the most common congenital craniofacial anomaly. Adverse healing following cleft palate repair can lead to oronasal fistula (ONF), a persistent connection between the oral and nasal cavities. Although human allograft tissues are currently the gold standard for ONF repair, these methods carry risks of infection and rejection, often requiring surgical revision. Immunoregenerative therapies present a novel alternative approach to harness the body's immune response and enhance the wound healing environment. An FDA-approved immunomodulatory drug, FTY720, is repurposed to reduce lymphocyte egress and induce immune cell fate switching toward pro-regenerative phenotypes. In this study, a bilayer biomaterial system is engineered using Tegaderm to secure and control the delivery of FTY720-nanofiber scaffolds (FTY720-NF). The release kinetics of the bilayer FTY720-NF is optimized to maintain drug release for up to 7 days, ensuring safe transdermal absorption and tissue biodistribution. The comprehensive immunophenotyping results demonstrate a regenerative state transition in hybrid immune cells recruited to the wound site. Further, histological evaluations reveal a significant ONF closure in mice by day 7 following bilayer FTY720-NF implantation. These findings demonstrate the utility of immunomodulatory strategies for oral wound healing, better positing the field to develop more efficacious treatment options in pediatric patients.
Collapse
Affiliation(s)
- Afra I Toma
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, 30322, USA
- Department of Pediatrics and Otolaryngology, Children's Healthcare of Atlanta, Atlanta, GA, 30329, USA
| | - Daniel Shah
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, 30322, USA
| | - Daniela Roth
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jeremie Oliver Piña
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lauren Hymel
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, 30322, USA
| | - Thomas Turner
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, 30322, USA
| | - Archana Kamalakar
- Department of Pediatrics and Otolaryngology, Children's Healthcare of Atlanta, Atlanta, GA, 30329, USA
| | - Ken Liu
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Perry Bartsch
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Leon Jacobs
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Rena D'Souza
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Dennis Liotta
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Edward Botchwey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, 30322, USA
| | - Nick J Willett
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, 97403, USA
| | - Steven L Goudy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, 30322, USA
- Department of Pediatrics and Otolaryngology, Children's Healthcare of Atlanta, Atlanta, GA, 30329, USA
| |
Collapse
|
6
|
Aghayants S, Zhu J, Yu J, Tao R, Li S, Zhou S, Zhou Y, Zhu Z. The emerging modulators of non-coding RNAs in diabetic wound healing. Front Endocrinol (Lausanne) 2024; 15:1465975. [PMID: 39439564 PMCID: PMC11493653 DOI: 10.3389/fendo.2024.1465975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Diabetic wound healing is a complex physiological process often hindered by the underlying metabolic dysfunctions associated with diabetes. Despite existing treatments, there remains a critical need to explore innovative therapeutic strategies to improve patient outcomes. This article comprehensively examines the roles of non-coding RNAs (ncRNAs), specifically microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), in regulating key phases of the wound healing process: inflammation, angiogenesis, re-epithelialization, and tissue remodeling. Through a deep review of current literature, we discuss recent discoveries of ncRNAs that have been shown to either promote or impair the wound healing process in diabetic wound healing, which were not covered in earlier reviews. This review highlights the specific mechanisms by which these ncRNAs impact cellular behaviors and pathways critical to each healing stage. Our findings indicate that understanding these recently identified ncRNAs provides new insights into their potential roles in diabetic wound healing, thereby contributing valuable knowledge for future research directions in this field.
Collapse
Affiliation(s)
- Sis Aghayants
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jinjin Zhu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Jing Yu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Tao
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Sicheng Li
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shengzhi Zhou
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yunhua Zhou
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhanyong Zhu
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
7
|
Kaussikaa S, Prasad MK, Ramkumar KM. Nrf2 Activation in Keratinocytes: A Central Role in Diabetes-Associated Wound Healing. Exp Dermatol 2024; 33:e15189. [PMID: 39373525 DOI: 10.1111/exd.15189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/28/2024] [Accepted: 09/21/2024] [Indexed: 10/08/2024]
Abstract
Wound healing is a complex biological process crucial for tissue repair, wherein keratinocytes play a pivotal role in initiating, sustaining and completing the cascade. Various local and systemic factors, such as lifestyle, age metabolic disorders and vascular insufficiency, can influence this process, and in the context of diabetic wounds, disrupted biological mechanisms, including inflammation, tissue hypoxia, decrease in collagen production along with increased oxidative stress and keratinocyte dysfunction, contribute to delayed healing. During re-epithelialisation, keratinocytes undergo rapid multiplication and migration, forming a dense hyperproliferative epithelial layer that restores the epidermal barrier. Nuclear factor-erythroid 2-related factor (Nrf2), a vital transcription factor, emerges as a central regulator in managing antioxidant proteins and detoxifying enzymes, serving as a guardian against elevated reactive oxygen species (ROS) levels during stress. Nrf2 also orchestrates angiogenesis and anti-inflammatory responses crucial for wound repair. Studies demonstrate that under high-glucose conditions, Nrf2 activation promotes wound healing by enhancing cell proliferation and migration while reducing apoptosis. Nrf2 activators stimulate endogenous antioxidant production, thereby mitigating oxidative stress. Furthermore, Nrf2 upregulation is associated with decreased expression of cytokines such as TNF-α and IL- 6. Recent research underscores the potential of bioactive molecules, including dietary polyphenols, traditional medicinal compounds and pharmacological agents, in activating Nrf2 and preventing diseases such as diabetes due to their robust antioxidative properties. This review aims to investigate the activation of Nrf2 by these bioactive molecules in cultured keratinocytes and animal models, elucidating the key molecular regulatory mechanisms involved in alleviating oxidative stress and facilitating the diabetic wound healing process. Understanding these complex pathways may offer insights into novel therapeutic strategies for enhanced wound healing in diabetes-associated complications.
Collapse
Affiliation(s)
- Srinivasan Kaussikaa
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Murali Krishna Prasad
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
8
|
Rahnama M, Ghasemzadeh N, Ebrahimi Y, Golchin A. A comprehensive evaluation of dermal fibroblast therapy in clinical trials for treating skin disorders and cosmetic applications: a scoping review. Stem Cell Res Ther 2024; 15:318. [PMID: 39304949 DOI: 10.1186/s13287-024-03892-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/25/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Fibroblast cells have the ability to improve skin conditions through regenerative medicine and cell-based therapies. The purpose of this scoping review is to assess the contribution of fibroblast cells to skin homeostasis and extracellular matrix deposition in clinical trials involving skin disorders and cosmetic applications. METHODS Using targeted search terms, published publications from January 2000 to August 2023 that addressed fibroblast uses in clinical trials of skin conditions were obtained from bibliographic databases like PubMed, Scopus, and Web of Science (WoS). Precise inclusion and exclusion criteria were used during the screening process. The potential benefits of induction treatment with fibroblasts lead to the choosing of clinical trials for this kind of treatment. RESULTS Out of the 820 published ppapers initially identified, only 35 studies fulfilled our meticulous eligibility criteria after careful screening. To ensure clarity, we methodically eliminated any duplicate or irrelevant published papers, thereby offering a transparent account of our selection process. CONCLUSION This study highlights the advantages of fibroblast therapy in treating skin conditions such as diabetic foot, venous leg ulcers, and cosmetic reasons. Fibroblasts possess remarkable regenerating capabilities, making dermal fibroblast therapy crucial in cell-based and skin regenerative treatments. Nevertheless, additional research is required for more disorders and cosmetic applications.
Collapse
Affiliation(s)
- Maryam Rahnama
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Navid Ghasemzadeh
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yaser Ebrahimi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Golchin
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran.
- Department of Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
9
|
He M, Niu J, Cheng H, Guo C. Identification and validation of diagnostic genes associated with neutrophil extracellular traps of type 2 diabetes mellitus. Front Genet 2024; 15:1373807. [PMID: 39296548 PMCID: PMC11408200 DOI: 10.3389/fgene.2024.1373807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 08/20/2024] [Indexed: 09/21/2024] Open
Abstract
Background Neutrophil extracellular traps (NETs) cause delayed wound closed up in type 2 diabetes mellitus (T2DM), but the specific regulatory mechanism of NETs-related genes (NETs-RGs) in T2DM is unclear. Methods We acquired GSE21321 and GSE15932 datasets from gene expression omnibus (GEO) database. First, differentially expressed genes (DEGs) between T2DM and control samples of GSE21321 dataset were sifted out by differential expression analysis. NETs scores were calculated for all samples in GSE21321 dataset, and key module genes associated with NETs scores were screened by constructing co-expression network. Then, DEGs and key module genes were intersected to yield intersection genes, and candidate genes were identified by constructing a protein protein interaction (PPI) network. Least absolute shrinkage and selection operator (LASSO) regression analysis was implemented on candidate genes to screen out diagnostic genes, and they were subjected to single sample gene set enrichment analysis (ssGSEA). Finally, immune characteristic analysis was carried out, and we constructed the gene-drug and transcription factor (TF)-miRNA-mRNA networks. Besides, we validated the expression of diagnostic genes by quantitative real-time polymerase chain reaction (qRT-PCR). Results In total, 23 candidate genes were gained by PPI analysis. The 5 diagnostic genes, namely, inter-trypsin inhibitor heavy chain 3 (ITIH3), fibroblast growth factor 1 (FGF1), neuron cell adhesion molecule (NRCAM), advanced glycosylation end-product-specific receptor (AGER), and calcium voltage-gated channel subunit alpha1 C (CACNA1C), were identified via LASSO analysis, and they were involved in carboxylic acid transport, axonogenesis, etc. M2 Macrophage, Monocyte, Natural killer (NK) cell, and Myeloid dendritic cells (DC) were remarkably different between T2DM and control samples. Diagnostic genes had the strongest and the most significant positive correlation with B cells. The gene-drug network included CACNA1C-Isradipine, CACNA1C-Benidipine and other relationship pairs. Totally 76 nodes and 44 edges constituted the TF-miRNA-mRNA network, including signal transducer and activator of transcription 1(STAT1) -hsa-miR-3170-AGER, CCCTC-binding factor (CTCF)-hsa-miR-455-5p-CACNA1C, etc. Moreover, qRT-PCR suggested that the expression trends of FGF1 and AGER were in keeping with the results of bioinformatic analysis. FGF1 and AGER were markedly regulated downwards in the T2DM group. Conclusion Through bioinformatic analysis, we identified NETs-related diagnostic genes (ITIH3, FGF1, NRCAM, AGER, CACNA1C) in T2DM, and explored their mechanism of action from different aspects, providing new ideas for the studies related to diagnosis and treatment of T2DM.
Collapse
Affiliation(s)
- Meifang He
- Endocrinoloy Department, Peking University First Hospital Taiyuan Hospital (Taiyuan Central Hospital), Taiyuan, China
| | - Jin Niu
- Endocrinoloy Department, Peking University First Hospital Taiyuan Hospital (Taiyuan Central Hospital), Taiyuan, China
| | - Haihua Cheng
- Endocrinoloy Department, Peking University First Hospital Taiyuan Hospital (Taiyuan Central Hospital), Taiyuan, China
| | - Chaoying Guo
- Endocrinoloy Department, Peking University First Hospital Taiyuan Hospital (Taiyuan Central Hospital), Taiyuan, China
| |
Collapse
|
10
|
Kartal B, Alimogullari E, Elçi P, Fatsa T, Ören S. The effects of Quercetin on wound healing in the human umbilical vein endothelial cells. Cell Tissue Bank 2024; 25:851-860. [PMID: 38944663 DOI: 10.1007/s10561-024-10144-1] [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: 12/10/2022] [Accepted: 06/12/2024] [Indexed: 07/01/2024]
Abstract
An injury that affects the integrity of the skin, either inside or externally, is called a wound. Damaged tissue is repaired by a set of cellular and molecular mechanisms known as wound healing. Quercetin, a naturally occurring flavonoid, may hasten the healing of wounds. The study's objective was to investigate any potential impacts of quercetin on the wound-healing process. Human umbilical vein endothelial cells (HUVECs) were treated to varying dose ranges of quercetin (5-320 nM) for 24 and 48 h. Cultured cells were evaluated by using the MTT analysis, wound scratch assay and vascular tube formation. Furthermore the gene expression of VEGF and FGF were evaluated by qRT-PCR to determine the effects of quercetin on angiogenezis and wound repair. Positive effects of quercetin on cellular viability were demonstrated by the MTT experiment. In HUVECs quercetin promoted tube formation, migration, and proliferation while also averting wound breakage. Moreover, quercetin increased the expression of the FGF and VEGF genes, which aid in the healing of wounds in HUVECs. Quercetin may be bioactive molecule that successfully speeds up wound healing by regulating the vasculogenezis and healing cells.
Collapse
Affiliation(s)
- Bahar Kartal
- Ankara Yıldırım Beyazıt Üniversitesi: Ankara Yildirim Beyazit Universitesi Ankara, Çankaya, Turkey.
| | - Ebru Alimogullari
- Ankara Yıldırım Beyazıt Üniversitesi: Ankara Yildirim Beyazit Universitesi Ankara, Çankaya, Turkey
| | - Pınar Elçi
- Ankara Yıldırım Beyazıt Üniversitesi: Ankara Yildirim Beyazit Universitesi Ankara, Çankaya, Turkey
| | - Tugba Fatsa
- Ankara Yıldırım Beyazıt Üniversitesi: Ankara Yildirim Beyazit Universitesi Ankara, Çankaya, Turkey
| | - Sema Ören
- Ankara Yıldırım Beyazıt Üniversitesi: Ankara Yildirim Beyazit Universitesi Ankara, Çankaya, Turkey
| |
Collapse
|
11
|
Smith J, Rai V. Novel Factors Regulating Proliferation, Migration, and Differentiation of Fibroblasts, Keratinocytes, and Vascular Smooth Muscle Cells during Wound Healing. Biomedicines 2024; 12:1939. [PMID: 39335453 PMCID: PMC11429312 DOI: 10.3390/biomedicines12091939] [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: 06/28/2024] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
Chronic diabetic foot ulcers (DFUs) are a significant complication of diabetes mellitus, often leading to amputation, increased morbidity, and a substantial financial burden. Even with the advancements in the treatment of DFU, the risk of amputation still exists, and this occurs due to the presence of gangrene and osteomyelitis. Nonhealing in a chronic DFU is due to decreased angiogenesis, granulation tissue formation, and extracellular matrix remodeling in the presence of persistent inflammation. During wound healing, the proliferation and migration of fibroblasts, smooth muscle cells, and keratinocytes play a critical role in extracellular matrix (ECM) remodeling, angiogenesis, and epithelialization. The molecular factors regulating the migration, proliferation, and differentiation of these cells are scarcely discussed in the literature. The literature review identifies the key factors influencing the proliferation, migration, and differentiation of fibroblasts, keratinocytes, and vascular smooth muscle cells (VSMCs), which are critical in wound healing. This is followed by a discussion on the various novel factors regulating the migration, proliferation, and differentiation of these cells but not in the context of wound healing; however, they may play a role. Using a network analysis, we examined the interactions between various factors, and the findings suggest that the novel factors identified may play a significant role in promoting angiogenesis, granulation tissue formation, and extracellular matrix remodeling during wound healing or DFU healing. However, these interactions warrant further investigation to establish their role alone or synergistically.
Collapse
Affiliation(s)
- Jacob Smith
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
12
|
Ren F, Wang L, Wang Y, Wang J, Wang Y, Song X, Zhang G, Nie F, Lin S. Single-cell transcriptome profiles the heterogeneity of tumor cells and microenvironments for different pathological endometrial cancer and identifies specific sensitive drugs. Cell Death Dis 2024; 15:571. [PMID: 39112478 PMCID: PMC11306564 DOI: 10.1038/s41419-024-06960-8] [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: 04/16/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
Endometrial cancer (EC) is a highly heterogeneous malignancy characterized by varied pathology and prognoses, and the heterogeneity of its cancer cells and the tumor microenvironment (TME) remains poorly understood. We conducted single-cell RNA sequencing (scRNA-seq) on 18 EC samples, encompassing various pathological types to delineate their specific unique transcriptional landscapes. Cancer cells from diverse pathological sources displayed distinct hallmarks labeled as immune-modulating, proliferation-modulating, and metabolism-modulating cancer cells in uterine clear cell carcinomas (UCCC), well-differentiated endometrioid endometrial carcinomas (EEC-I), and uterine serous carcinomas (USC), respectively. Cancer cells from the UCCC exhibited the greatest heterogeneity. We also identified potential effective drugs and confirmed their effectiveness using patient-derived EC organoids for each pathological group. Regarding the TME, we observed that prognostically favorable CD8+ Tcyto and NK cells were prominent in normal endometrium, whereas CD4+ Treg, CD4+ Tex, and CD8+ Tex cells dominated the tumors. CXCL3+ macrophages associated with M2 signature and angiogenesis were exclusively found in tumors. Prognostically relevant epithelium-specific cancer-associated fibroblasts (eCAFs) and SOD2+ inflammatory CAFs (iCAFs) predominated in EEC-I and UCCC groups, respectively. We also validated the oncogenic effects of SOD2+ iCAFs in vitro. Our comprehensive study has yielded deeper insights into the pathogenesis of EC, potentially facilitating personalized treatments for its varied pathological types.
Collapse
Affiliation(s)
- Fang Ren
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Lingfang Wang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuyouye Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiaxuan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuanpei Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaole Song
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Gong Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fangfang Nie
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shitong Lin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, 430022, Wuhan, Hubei, PR China.
| |
Collapse
|
13
|
Shen W, Mao Y, Ge X, Xu J, Hu J, Ao F, Wu S, Yan P. PLA tissue-engineered scaffolds loaded with sustained-release active substance chitosan nanoparticles: Modeling BSA-bFGF as the active substance. Int J Biol Macromol 2024; 274:133120. [PMID: 38876244 DOI: 10.1016/j.ijbiomac.2024.133120] [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: 10/06/2023] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
The utilization of basic fibroblast growth factor (bFGF) in the development of tissue-engineered scaffolds is both challenging and imperative. In our pursuit of creating a scaffold that aligns with the natural healing process, we initially fabricated chitosan-bFGF nanoparticles (CS-bFGF NPs) through electrostatic spraying. Subsequently, polylactic acid (PLA) fiber was prepared using electrospinning technique, and the CS-bFGF NPs were uniformly embedded within the pores of porous PLA fibers. Scanning electron micrographs illustrate the smooth surface of the nanoparticles, showing a porous structure intricately attached to PLA fibers. Fourier-transform infrared spectroscopy (FTIR), energy-dispersive X-ray spectroscopy (EDS), and X-ray diffraction (XRD) analyses provided conclusive evidence that the CS-bFGF NPs were uniformly distributed throughout the porous PLA fibers, forming a robust physical bond through electrostatic adsorption. The resultant scaffolds exhibited commendable mechanical properties and hydrophilicity, facilitating a sustained-release for 72 h. Furthermore, the biocompatibility and degradation performance of the scaffolds were substantiated by monitoring conductivity and pH changes in pure water over different time intervals, complemented by scanning electron microscopy (SEM) observations. Cell experiments confirmed the cytocompatibility of the scaffolds. In animal studies, the group treated with 16 % NPs/Scaffold demonstrated the highest epidermal reconstruction rate. In summary, our developed materials present a promising candidate for serving as a tissue engineering scaffold, showcasing exceptional biocompatibility, sustained-release characteristics, and substantial potential for promoting epidermal regeneration.
Collapse
Affiliation(s)
- Wen Shen
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Yueyang Mao
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Xuemei Ge
- College of Light Industry and Food Engineering, Nanjing Forestry University Nanjing, Nan Jing 210037, China
| | - Jingwen Xu
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Jiaru Hu
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Fen Ao
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Shang Wu
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Pi Yan
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| |
Collapse
|
14
|
Yadav JP, Verma A, Pathak P, Dwivedi AR, Singh AK, Kumar P, Khalilullah H, Jaremko M, Emwas AH, Patel DK. Phytoconstituents as modulators of NF-κB signalling: Investigating therapeutic potential for diabetic wound healing. Biomed Pharmacother 2024; 177:117058. [PMID: 38968797 DOI: 10.1016/j.biopha.2024.117058] [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: 04/15/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/07/2024] Open
Abstract
The NF-κB pathway plays a pivotal role in impeding the diabetic wound healing process, contributing to prolonged inflammation, diminished angiogenesis, and reduced proliferation. In contrast to modern synthetic therapies, naturally occurring phytoconstituents are well-studied inhibitors of the NF-κB pathway that are now attracting increased attention in the context of diabetic wound healing because of lower toxicity, better safety and efficacy, and cost-effectiveness. This study explores recent research on phytoconstituent-based therapies and delve into their action mechanisms targeting the NF-κB pathway and potential for assisting effective healing of diabetic wounds. For this purpose, we have carried out surveys of recent literature and analyzed studies from prominent databases such as Science Direct, Scopus, PubMed, Google Scholar, EMBASE, and Web of Science. The classification of phytoconstituents into various categorie such as: alkaloids, triterpenoids, phenolics, polyphenols, flavonoids, monoterpene glycosides, naphthoquinones and tocopherols. Noteworthy phytoconstituents, including Neferine, Plumbagin, Boswellic acid, Genistein, Luteolin, Kirenol, Rutin, Vicenin-2, Gamma-tocopherol, Icariin, Resveratrol, Mangiferin, Betulinic acid, Berberine, Syringic acid, Gallocatechin, Curcumin, Loureirin-A, Loureirin-B, Lupeol, Paeoniflorin, and Puerarin emerge from these studies as promising agents for diabetic wound healing through the inhibition of the NF-κB pathway. Extensive research on various phytoconstituents has revealed how they modulate signalling pathways, including NF-κB, studies that demonstrate the potential for development of therapeutic phytoconstituents to assist healing of chronic diabetic wounds.
Collapse
Affiliation(s)
- Jagat Pal Yadav
- Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India; Pharmacology Research Laboratory, Faculty of Pharmaceutical Sciences, Rama University, Kanpur 209217, India; Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India.
| | - Amita Verma
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India
| | - Prateek Pathak
- Department of Pharmaceutical Analysis, Quality Assurance and Pharmaceutical Chemistry, GITAM School of Pharmacy, GITAM (Deemed to be University), Hyderabad Campus, 502329, India
| | - Ashish R Dwivedi
- Department of Pharmaceutical Analysis, Quality Assurance and Pharmaceutical Chemistry, GITAM School of Pharmacy, GITAM (Deemed to be University), Hyderabad Campus, 502329, India
| | - Ankit Kumar Singh
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India; Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda 151401, India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda 151401, India
| | - Habibullah Khalilullah
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Unayzah 51911, Saudi Arabia
| | - Mariusz Jaremko
- Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Abdul-Hamid Emwas
- Core Labs, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Dinesh Kumar Patel
- Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India.
| |
Collapse
|
15
|
Guo L, Xiao D, Xing H, Yang G, Yang X. Engineered exosomes as a prospective therapy for diabetic foot ulcers. BURNS & TRAUMA 2024; 12:tkae023. [PMID: 39026930 PMCID: PMC11255484 DOI: 10.1093/burnst/tkae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/29/2024] [Indexed: 07/20/2024]
Abstract
Diabetic foot ulcer (DFU), characterized by high recurrence rate, amputations and mortality, poses a significant challenge in diabetes management. The complex pathology involves dysregulated glucose homeostasis leading to systemic and local microenvironmental complications, including peripheral neuropathy, micro- and macro-angiopathy, recurrent infection, persistent inflammation and dysregulated re-epithelialization. Novel approaches to accelerate DFU healing are actively pursued, with a focus on utilizing exosomes. Exosomes are natural nanovesicles mediating cellular communication and containing diverse functional molecular cargos, including DNA, mRNA, microRNA (miRNA), lncRNA, proteins, lipids and metabolites. While some exosomes show promise in modulating cellular function and promoting ulcer healing, their efficacy is limited by low yield, impurities, low loading content and inadequate targeting. Engineering exosomes to enhance their curative activity represents a potentially more efficient approach for DFUs. This could facilitate focused repair and regeneration of nerves, blood vessels and soft tissue after ulcer development. This review provides an overview of DFU pathogenesis, strategies for exosome engineering and the targeted therapeutic application of engineered exosomes in addressing critical pathological changes associated with DFUs.
Collapse
Affiliation(s)
- Lifei Guo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an 710032, China
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an 710032, China
- Cadet Team 6 of School of Basic Medicine, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an 710032, China
| | - Dan Xiao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an 710032, China
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an 710032, China
| | - Helin Xing
- Department of Prosthodontics, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Tiantanxili Street #4, Dongcheng District, Beijing 100050, China
| | - Guodong Yang
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an 710032, China
| | - Xuekang Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi'an 710032, China
| |
Collapse
|
16
|
Wong AYW, Hooi NMF, Yeo BSY, Sultana R, Bee YM, Lee ARYB, Tay SM. Improving Diabetic Wound-Healing Outcomes With Topical Growth Factor Therapies. J Clin Endocrinol Metab 2024; 109:e1642-e1651. [PMID: 38477463 DOI: 10.1210/clinem/dgae128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024]
Abstract
CONTEXT Diabetes mellitus is associated with morbid complications such as diabetic foot ulcers (DFUs) that may lead to amputations or mortality if not managed adequately. OBJECTIVE New adjunctive interventions to treat diabetic wounds include topical biologics and growth factors. This study aims to evaluate their efficacy in improving wound-healing outcomes and safety. METHODS Comprehensive database searches of MEDLINE via PubMed, EMBASE, and Cochrane were performed from inception to December 2022. Three independent researchers selected the studies. Randomized controlled trials that compared the use of a topical biologic growth factor-containing regimen to other biologics or standard of care (SOC) were included. This review followed PRISMA guidelines. Risk of bias analysis was performed using the Jadad scale. Network meta-analysis was performed. Treatments were grouped into common nodes based on the type of biologic agent. Primary outcomes of interest were healing rate and time to wound closure. Secondary outcomes included wound infection, serious adverse events (AEs), and amputation rate. RESULTS Human umbilical cord (HUC) was associated with the highest cure, followed by recombinant human epidermal growth factor (hEGF). A significantly greater reduction in the time to cure DFUs was seen in HUC, hEGF, and fibroblast growth factor (FGF). There was a significantly lower risk of AEs when platelet-rich plasma (PRP) was administered. CONCLUSION HUC, hEGF, and FGF are promising topical biologics with statistically significant primary outcomes compared to SOC, while PRP is effective in reducing ulcer-related AEs. HUC has been found to be the most effective in terms of cure rate and a reduction in time to cure.
Collapse
Affiliation(s)
- Andrew Yew Wei Wong
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Natalie Ming Foong Hooi
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Brian Sheng Yep Yeo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Rehena Sultana
- Duke-NUS Medical School, National University of Singapore, Singapore 169857, Singapore
| | - Yong Mong Bee
- Department of Endocrinology, Singapore General Hospital, Singapore 169608, Singapore
| | - Ainsley Ryan Yan Bin Lee
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Sook Muay Tay
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Department of Surgical Intensive Care, Singapore General Hospital, Singapore 169608, Singapore
| |
Collapse
|
17
|
Wang L, Zhang X, Li H, Mou Y, Cui G. SP1 promotes high glucose-induced lens epithelial cell viability, migration and epithelial-mesenchymal transition via regulating FGF7 and PI3K/AKT pathway. Int Ophthalmol 2024; 44:316. [PMID: 38969958 DOI: 10.1007/s10792-024-03230-6] [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: 01/23/2024] [Accepted: 06/18/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Diabetic cataract (DC) is a common complication of diabetes and its etiology and progression are multi-factorial. In this study, the roles of specific protein 1 (SP1) and fibroblast growth factor 7 (FGF7) in DC development were explored. METHODS DC cell model was established by treating SRA01/04 cells with high glucose (HG). MTT assay was conducted to evaluate cell viability. Transwell assay and wound-healing assay were performed to assess cell migration and invasion. Western blot assay and qRT-PCR assay were conducted to measure the expression of N-cadherin, E-cadherin, Collagen I, Fibronectin, SP1 and FGF7 expression. CHIP assay and dual-luciferase reporter assay were conducted to analyze the combination between FGF7 and SP1. RESULTS FGF7 was upregulated in DC patients and HG-induced SRA01/04 cells. HG treatment promoted SRA01/04 cell viability, migration, invasion and epithelial-mesenchymal transition (EMT), while FGF7 knockdown abated the effects. Transcription factor SP1 activated the transcription level of FGF7 and SP1 overexpression aggravated HG-induced SRA01/04 cell injury. SP1 silencing repressed HG-induced SRA01/04 cell viability, migration, invasion and EMT, but these effects were ameliorated by upregulating FGF7. Additionally, SP1 knockdown inhibited the PI3K/AKT pathway by regulating the transcription level of FGF7. CONCLUSION Transcription factor SP1 activated the transcription level of FGF7 and the PI3K/AKT pathway to regulate HG-induced SRA01/04 cell viability, migration, invasion and EMT.
Collapse
Affiliation(s)
- Ledan Wang
- Department of Ophthalmology, Ophthalmology Center, Taizhou Hospital of Zhejiang Province, 150 Ximen Street, Linhai, 317000, China
| | - Xin Zhang
- Department of Ophthalmology, Ophthalmology Center, Taizhou Hospital of Zhejiang Province, 150 Ximen Street, Linhai, 317000, China
| | - Huijun Li
- Department of Ophthalmology, Ophthalmology Center, Taizhou Hospital of Zhejiang Province, 150 Ximen Street, Linhai, 317000, China
| | - Yuehong Mou
- Department of Ophthalmology, Ophthalmology Center, Taizhou Hospital of Zhejiang Province, 150 Ximen Street, Linhai, 317000, China
| | - Gangfeng Cui
- Department of Ophthalmology, Ophthalmology Center, Taizhou Hospital of Zhejiang Province, 150 Ximen Street, Linhai, 317000, China.
| |
Collapse
|
18
|
Wang G, Li X, Ju S, Li Y, Li W, He H, Cai Y, Dong Z, Fu W. Effect of electrospun poly (L-lactide-co-caprolactone) and formulated porcine fibrinogen for diabetic foot ulcers. Eur J Pharm Sci 2024; 198:106800. [PMID: 38754593 DOI: 10.1016/j.ejps.2024.106800] [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: 03/23/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
Diabetic foot ulcers were a significant complication of diabetes and were accompanied by delayed wound healing. To compare the effect of topical application electrospun poly (L-lactide-co-caprolactone) and formulated porcine fibrinogen (PLCL/Fg) dressing with alginate dressing when treating diabetic foot ulcers (DFUs). A single-center, prospective, randomized, patient-blinded clinical trial was conducted from July 1, 2023, to December 26, 2023. The clinical trial registration was completed on August 28, 2023 (ClinicalTrials.gov Identifier: NCT06014437). The eligible patients with DFUs of 1-20 cm2 present for at least 1 month and with Wagner grade 1 or 2. They were randomized 1:1 to receive PLCL/Fg or alginate dressing. Participants received PLCL/Fg dressing 1-3 times per week or alginate dressing 3 times per week for 12 weeks. A total of 52 patients (33 men [63.5 %]; mean [SD] age, 63.1 [11.9] years; mean [SD] diabetes time, 8.3 [4.6] years) with DFUs were assessed for this study. The DFUs classified as Wagner grade 1 or 2 (mean [SD] ulcer area, 3.8 [3.2] cm2) were randomized to receive either the PLCL/Fg dressing (n = 26) or the alginate dressing (n = 26) for as long as 12 weeks. In this study, the incidence of complete healing included 22 patients (91.7 %) in the PLCL/Fg group and 14 (63.6 %) in the alginate group during the 12-week treatment period (P = 0.003). The treatment-related adverse events that occurred were 5 (20.8 %) in the PLCL/Fg group and 4 (18.1 %) in the comparator group. In this randomized clinical trial, PLCL/Fg dressing showed beneficial effects in DFUs treatment of wound surface reduction and regulating the wound microenvironment.
Collapse
Affiliation(s)
- Guili Wang
- Department of Vascular and wound center, Jinshan Hospital, Fudan University, Shanghai 200032, China; Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Vascular Surgery Institute of Fudan University, Shanghai 200032, China
| | - Xiaoyan Li
- Department of Vascular and wound center, Jinshan Hospital, Fudan University, Shanghai 200032, China
| | - Shuai Ju
- Department of Vascular and wound center, Jinshan Hospital, Fudan University, Shanghai 200032, China
| | - Yao Li
- Department of Vascular and wound center, Jinshan Hospital, Fudan University, Shanghai 200032, China
| | - Wenqiang Li
- Department of Vascular and wound center, Jinshan Hospital, Fudan University, Shanghai 200032, China
| | - Hongbing He
- PINE&POWER Biotech Co., Ltd, Shanghai, China; Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Vascular Surgery Institute of Fudan University, Shanghai 200032, China
| | - Yunmin Cai
- Department of Vascular and wound center, Jinshan Hospital, Fudan University, Shanghai 200032, China.
| | - Zhihui Dong
- Department of Vascular and wound center, Jinshan Hospital, Fudan University, Shanghai 200032, China; Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Vascular Surgery Institute of Fudan University, Shanghai 200032, China.
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Vascular Surgery Institute of Fudan University, Shanghai 200032, China
| |
Collapse
|
19
|
Zhao F, Xie L, Weng Z, Huang Y, Zheng L, Yan S, Shen X. Combined with dynamic serum proteomics and clinical follow-up to screen the serum proteins to promote the healing of diabetic foot ulcer. Endocrine 2024; 84:365-379. [PMID: 37938414 DOI: 10.1007/s12020-023-03579-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/17/2023] [Indexed: 11/09/2023]
Abstract
OBJECTIVE Non-healing diabetic foot ulcers are a leading cause of disability and death in diabetic patients, which often results in lower limb amputation. This study aimed to investigate the impact of biomarkers on the healing of diabetic foot ulcers by utilizing dynamic serum proteomics and skin proteomic analysis, combined with clinical case follow-up studies. METHODS To analyze dynamic serum proteomic changes in four groups, age-matched normal subjects, diabetic patients, pretreatment diabetic foot ulcer patients, and healed diabetic foot ulcer patients were selected. The differential proteins were screened in conjunction with normal and diabetic foot ulcer skin proteomics. In this study, a total of 80 patients with diabetic foot ulcers were enrolled and monitored for 3-6 months during treatment. To verify the significance of the differential proteins, age-matched diabetic patients (240 patients) and healthy controls (160 patients) were included as controls. RESULTS Dynamic serum proteomics trend showed that the level of negative regulatory proteins related to endothelial cell migration, angiogenesis, and vascular development was significantly decreased after treatment of diabetic foot ulcer. GO enrichment analysis suggested that differentially expressed proteins were mainly enriched in protein activation cascade, immunoglobulin production, and complement activation. The researchers identified the core proteins APOA1, LPA, and APOA2 through a convergence of serum and skin proteomics screening. Clinical cases further validated that APOA1 levels are decreased in diabetic foot ulcer patients and are correlated with disease severity. In addition, animal experiments showed that APOA1 could promote wound healing in diabetic mice. CONCLUSIONS Based on our dynamic proteomics and clinical case studies, our bioinformatic analysis suggests that APOA1 plays a critical role in linking coagulation, inflammation, angiogenesis, and wound repair, making it a key protein that promotes the healing of diabetic foot ulcers.
Collapse
Affiliation(s)
- Fengying Zhao
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Endocrinology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Diabetes Research Institute of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Metabolic Diseases Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Liangxiao Xie
- Department of Endocrinology and Metabolism, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, 363000, China
| | - Zhiyan Weng
- Department of Endocrinology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Diabetes Research Institute of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Metabolic Diseases Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Yihong Huang
- Department of Endocrinology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Diabetes Research Institute of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Metabolic Diseases Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Lifeng Zheng
- Orthopedics Department, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Sunjie Yan
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Endocrinology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
- Clinical Research Center for Metabolic Diseases of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Diabetes Research Institute of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Metabolic Diseases Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Ximei Shen
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Endocrinology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
- Clinical Research Center for Metabolic Diseases of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Diabetes Research Institute of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Metabolic Diseases Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
20
|
Tian H, Yu JL, Chu X, Guan Q, Liu J, Liu Y. Unraveling the role of C1GALT1 in abnormal glycosylation and colorectal cancer progression. Front Oncol 2024; 14:1389713. [PMID: 38699634 PMCID: PMC11063370 DOI: 10.3389/fonc.2024.1389713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 03/25/2024] [Indexed: 05/05/2024] Open
Abstract
C1GALT1 plays a pivotal role in colorectal cancer (CRC) development and progression through its involvement in various molecular mechanisms. This enzyme is central to the O-glycosylation process, producing tumor-associated carbohydrate antigens (TACA) like Tn and sTn, which are linked to cancer metastasis and poor prognosis. The interaction between C1GALT1 and core 3 synthase is crucial for the synthesis of core 3 O-glycans, essential for gastrointestinal health and mucosal barrier integrity. Aberrations in this pathway can lead to CRC development. Furthermore, C1GALT1's function is significantly influenced by its molecular chaperone, Cosmc, which is necessary for the proper folding of T-synthase. Dysregulation in this complex interaction contributes to abnormal O-glycan regulation, facilitating cancer progression. Moreover, C1GALT1 affects downstream signaling pathways and cellular behaviors, such as the epithelial-mesenchymal transition (EMT), by modifying O-glycans on key receptors like FGFR2, enhancing cancer cell invasiveness and metastatic potential. Additionally, the enzyme's relationship with MUC1, a mucin protein with abnormal glycosylation in CRC, highlights its role in cancer cell immune evasion and metastasis. Given these insights, targeting C1GALT1 presents a promising therapeutic strategy for CRC, necessitating further research to develop targeted inhibitors or activators. Future efforts should also explore C1GALT1's potential as a biomarker for early diagnosis, prognosis, and treatment response monitoring in CRC, alongside investigating combination therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Hong Tian
- Department of Oncology, Fourth People’s Hospital in Shenyang, China Medical University, Shenyang, China
| | - Jia-Li Yu
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Xiaoli Chu
- Department of Oncology, Fourth People’s Hospital in Shenyang, China Medical University, Shenyang, China
| | - Qi Guan
- Department of Oncology, Fourth People’s Hospital in Shenyang, China Medical University, Shenyang, China
| | - Juan Liu
- Department of Oncology, Fourth People’s Hospital in Shenyang, China Medical University, Shenyang, China
| | - Ying Liu
- Department of Oncology, Fourth People’s Hospital in Shenyang, China Medical University, Shenyang, China
| |
Collapse
|
21
|
Jin N, Wu J, Ye S, Xue J, Meng T, Hu L, Jiang S, Xu H, Yu Y, Hu D, Zhang G. Injectable Dynamic ROS-Responsive COF-Modified Microalgae Gels for In Vivo bFGF Delivery to Treat Diabetic Wounds. ACS APPLIED MATERIALS & INTERFACES 2024; 16:18608-18626. [PMID: 38565551 DOI: 10.1021/acsami.4c01509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Hypoxia, chronic inflammation, and elevated reactive oxygen species (ROS) production induced by hyperglycemia pose formidable challenges to the healing of diabetic chronic wounds, often resulting in impaired recovery. Currently, sustainable and eco-friendly therapeutic approaches targeting this multifaceted problem remain uncharted. Herein, we develop a unique three-functional covalent organic framework (COF)-modified microalgae gel designed for the preparation and treatment of chronic diabetic wounds. The gel comprises an oxygen-releasing basic fibroblast growth factor (bFGF) microalgae matrix, augmented by an ROS-responsive COF. Although two of these components have been reported to be used in wound healing, the combination of all three functions represents an innovative approach to synergize the treatment of chronic diabetic wounds. Therefore, we propose a new concept of "ligand interlocking" with three functional synergistic effects. Specifically, the COF has a similar effect to the "double Excalibur", which binds bFGF to promote angiogenesis and proliferation and inhibit the inflammatory response of chronic wounds and binds live microalgae to eliminate ROS and release dissolved oxygen to alleviate the hypoxia of wounds. Moreover, in vivo experiments and RNA sequencing analyses similarly demonstrated that the COF-modified microalgae gel reduced the inflammatory cascade cycle in the wound site and promoted vascular and tissue regeneration. We posit that the COF-modified microalgae gel represents a promising strategy for the active in vivo delivery of therapeutics to the wound body in intensive care unit settings.
Collapse
Affiliation(s)
- Neng Jin
- Department of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Jin Wu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Shuming Ye
- Department of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Jing Xue
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Tao Meng
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Liang Hu
- Department of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Shangshang Jiang
- Department of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Hanyang Xu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Yue Yu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Danyou Hu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Guiyang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
22
|
Sousa LPB, Pinto LFB, Cruz VAR, Oliveira GA, Rojas de Oliveira H, Chud TS, Pedrosa VB, Miglior F, Schenkel FS, Brito LF. Genome-wide association and functional genomic analyses for various hoof health traits in North American Holstein cattle. J Dairy Sci 2024; 107:2207-2230. [PMID: 37939841 DOI: 10.3168/jds.2023-23806] [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: 05/28/2023] [Accepted: 10/19/2023] [Indexed: 11/10/2023]
Abstract
Hoof diseases are a major welfare and economic issue in the global dairy cattle production industry, which can be minimized through improved management and breeding practices. Optimal genetic improvement of hoof health could benefit from a deep understanding of the genetic background and biological underpinning of indicators of hoof health. Therefore, the primary objectives of this study were to perform genome-wide association studies, using imputed high-density genetic markers data from North American Holstein cattle, for 8 hoof-related traits: digital dermatitis, sole ulcer, sole hemorrhage, white line lesion, heel horn erosion, interdigital dermatitis, interdigital hyperplasia, and toe ulcer, and a hoof health index. De-regressed estimated breeding values from 25,580 Holstein animals were used as pseudo-phenotypes for the association analyses. The genomic quality control, genotype phasing, and genotype imputation were performed using the PLINK (version 1.9), Eagle (version 2.4.1), and Minimac4 software, respectively. The functional genomic analyses were performed using the GALLO R package and the DAVID platform. We identified 22, 34, 14, 22, 28, 33, 24, 43, and 15 significant markers for digital dermatitis, heel horn erosion, interdigital dermatitis, interdigital hyperplasia, sole hemorrhage, sole ulcer, toe ulcer, white line lesion disease, and the hoof health index, respectively. The significant markers were located across all autosomes, except BTA10, BTA12, BTA20, BTA26, BTA27, and BTA28. Moreover, the genomic regions identified overlap with various previously reported quantitative trait loci for exterior, health, meat and carcass, milk, production, and reproduction traits. The enrichment analyses identified 44 significant gene ontology terms. These enriched genomic regions harbor various candidate genes previously associated with bone development, metabolism, and infectious and immunological diseases. These findings indicate that hoof health traits are highly polygenic and influenced by a wide range of biological processes.
Collapse
Affiliation(s)
- Luis Paulo B Sousa
- Department of Animal Sciences, Federal University of Bahia, Salvador, BA, 40170-110, Brazil
| | - Luis Fernando B Pinto
- Department of Animal Sciences, Federal University of Bahia, Salvador, BA, 40170-110, Brazil
| | - Valdecy A R Cruz
- Department of Animal Sciences, Federal University of Bahia, Salvador, BA, 40170-110, Brazil
| | - Gerson A Oliveira
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Hinayah Rojas de Oliveira
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada; Department of Animal Sciences, Purdue University, West Lafayette, IN 47907
| | - Tatiane S Chud
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada; PEAK, Madison, WI 53718
| | - Victor B Pedrosa
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907
| | - Filippo Miglior
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada; Lactanet Canada, Guelph, ON, N1K 1E5, Canada
| | - Flávio S Schenkel
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Luiz F Brito
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada; Department of Animal Sciences, Purdue University, West Lafayette, IN 47907.
| |
Collapse
|
23
|
Esmaeilzadeh A, Yeganeh PM, Nazari M, Esmaeilzadeh K. Platelet-derived extracellular vesicles: a new-generation nanostructured tool for chronic wound healing. Nanomedicine (Lond) 2024; 19:915-941. [PMID: 38445377 DOI: 10.2217/nnm-2023-0344] [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: 03/07/2024] Open
Abstract
Chronic nonhealing wounds pose a serious challenge to regaining skin function and integrity. Platelet-derived extracellular vesicles (PEVs) are nanostructured particles with the potential to promote wound healing since they can enhance neovascularization and cell migration and reduce inflammation and scarring. This work provides an innovative overview of the technical laboratory issues in PEV production, PEVs' role in chronic wound healing and the benefits and challenges in its clinical translation. The article also explores the challenges of proper sourcing, extraction techniques and storage conditions, and discusses the necessity of further evaluations and combinational therapeutics, including dressing biomaterials, M2-derived exosomes, mesenchymal stem cells-derived extracellular vesicles and microneedle technology, to boost their therapeutic efficacy as advanced strategies for wound healing.
Collapse
Affiliation(s)
- Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, 77978-45157, Iran
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, 77978-45157, Iran
| | | | - Mahdis Nazari
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, 77978-45157, Iran
| | - Kimia Esmaeilzadeh
- Department of Medical Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, 77978-45157, Iran
| |
Collapse
|
24
|
Li Y, Leng Y, Liu Y, Zhong J, Li J, Zhang S, Li Z, Yang K, Kong X, Lao W, Bi C, Zhai A. Advanced multifunctional hydrogels for diabetic foot ulcer healing: Active substances and biological functions. J Diabetes 2024; 16:e13537. [PMID: 38599855 PMCID: PMC11006623 DOI: 10.1111/1753-0407.13537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/26/2023] [Accepted: 01/18/2024] [Indexed: 04/12/2024] Open
Abstract
AIM Hydrogels with excellent biocompatibility and biodegradability can be used as the desirable dressings for the therapy of diabetic foot ulcer (DFU). This review aimed to summarize the biological functions of hydrogels, combining with the pathogenesis of DFU. METHODS The studies in the last 10 years were searched and summarized from the online database PubMed using a combination of keywords such as hydrogel and diabetes. The biological functions of hydrogels and their healing mechanism on DFU were elaborated. RESULTS In this review, hydrogels were classified by their active substances such as drugs, cytokines, photosensitizers, and biomimetic peptide. Based on this, the biological functions of hydrogels were summarized by associating the pathogenesis of DFU, including oxidative stress, chronic inflammation, cell phenotype change, vasculopathy, and infection. This review also pointed out some of the shortcomings of hydrogels in present researches. CONCLUSIONS Hydrogels were classified into carrier hydrogels and self-functioning hydrogels in this review. Besides, the functions and components of existing hydrogels were clarified to provide assistance for future researches and clinical applications.
Collapse
Affiliation(s)
- Yuetong Li
- Department of Endocrinology, The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Yuxin Leng
- Department of Critical Care MedicinePeking University Third HospitalBeijingChina
| | - Yang Liu
- Department of Laboratory Medicine, The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Jianhua Zhong
- Department of Endocrinology, The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Jiaxin Li
- Department of Endocrinology, The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Shitong Zhang
- Department of General Practice, The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Zhenlin Li
- Department of Endocrinology, The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Kaming Yang
- Department of Endocrinology, The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Xinyi Kong
- Department of Laboratory Medicine, The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Wanwen Lao
- Department of Endocrinology, The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Changlong Bi
- Department of Endocrinology, The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Aixia Zhai
- Department of Laboratory Medicine, The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| |
Collapse
|
25
|
Benchaprathanphorn K, Muangman P, Chinaroonchai K, Namviriyachote N, Ampawong S, Angkhasirisap W, Kengkoom K, Viravaidya-Pasuwat K. Translational application of human keratinocyte-fibroblast cell sheets for accelerated wound healing in a clinically relevant type 2 diabetic rat model. Cytotherapy 2024; 26:360-371. [PMID: 38363247 DOI: 10.1016/j.jcyt.2024.01.003] [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: 08/09/2023] [Revised: 12/27/2023] [Accepted: 01/20/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND AIMS Despite advancements in wound care, wound healing remains a challenge, especially in individuals with type 2 diabetes. Cell sheet technology has emerged as an efficient and promising therapy for tissue regeneration and wound repair. Among these, bilayered human keratinocyte-fibroblast cell sheets constructed using temperature-responsive culture surfaces have been shown to mimic a normal tissue-like structure and secrete essential cytokines and growth factors that regulate the wound healing process. METHODS This study aimed to evaluate the safety and therapeutic potential of human skin cell sheets to treat full-thickness skin defects in a rat model of type 2 diabetes. RESULTS Our findings demonstrate that diabetic wounds transplanted with bilayered cell sheets resulted in accelerated re-epithelialization, increased angiogenesis, enhanced macrophage polarization and regeneration of tissue that closely resembled healthy skin. In contrast, the control group that did not receive cell sheet transplantation presented characteristic symptoms of impaired and delayed wound healing associated with type 2 diabetes. CONCLUSIONS The secretory cytokines and the upregulation of Nrf2 expression in response to cell sheet transplantation are believed to have played a key role in the improved wound healing observed in diabetic rats. Our study suggests that human keratinocyte-fibroblast cell sheets hold great potential as a therapeutic alternative for diabetic ulcers.
Collapse
Affiliation(s)
- Kanokaon Benchaprathanphorn
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Pornprom Muangman
- Trauma Surgery Division, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kusuma Chinaroonchai
- Trauma Surgery Division, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nantaporn Namviriyachote
- Trauma Surgery Division, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wannee Angkhasirisap
- Research and Academic Support Office, National Laboratory Animal Center, Mahidol University, Nakorn Pathom, Thailand
| | - Kanchana Kengkoom
- Research and Academic Support Office, National Laboratory Animal Center, Mahidol University, Nakorn Pathom, Thailand
| | - Kwanchanok Viravaidya-Pasuwat
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand; Chemical Engineering, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand.
| |
Collapse
|
26
|
Cecerska-Heryć E, Goszka M, Gliźniewicz M, Grygorcewicz B, Serwin N, Stodolak P, Słodzińska W, Birger R, Polikowska A, Budkowska M, Rakoczy R, Dołęgowska B. The Effect of a Rotating Magnetic Field on the Regenerative Potential of Platelets. Int J Mol Sci 2024; 25:3644. [PMID: 38612456 PMCID: PMC11012199 DOI: 10.3390/ijms25073644] [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: 01/25/2024] [Revised: 02/25/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Platelets are actively involved in tissue injury site regeneration by producing a wide spectrum of platelet-derived growth factors such as PDGF (platelet-derived growth factor), IGF-1 (insulin-like growth factor), TGF-β1 (transforming growth factor β), FGF (fibroblast growth factor), etc. A rotating magnetic field (RMF) can regulate biological functions, including reduction or induction regarding inflammatory processes, cell differentiation, and gene expression, to determine the effect of an RMF on the regenerative potential of platelets. The study group consisted of 30 healthy female and male volunteers (n = 15), from which plasma was collected. A portion of the plasma was extracted and treated as an internal control group. Subsequent doses of plasma were exposed to RMF at different frequencies (25 and 50 Hz) for 1 and 3 h. Then, the concentrations of growth factors (IGF-1, PDGF-BB, TGF-β1, and FGF-1) were determined in the obtained material by the ELISA method. There were statistically significant differences in the PDGF-BB, TGF-β1, IGF-1, and FGF-1 concentrations between the analyzed groups. The highest concentration of PDGF-BB was observed in the samples placed in RMF for 1 h at 25 Hz. For TGF-β1, the highest concentrations were obtained in the samples exposed to RMF for 3 h at 25 Hz and 1 h at 50 Hz. The highest concentrations of IGF-1 and FGF-1 were shown in plasma placed in RMF for 3 h at 25 Hz. An RMF may increase the regenerative potential of platelets. It was noted that female platelets may respond more strongly to RMF than male platelets.
Collapse
Affiliation(s)
- Elżbieta Cecerska-Heryć
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (M.G.); (N.S.); (P.S.); (W.S.); (R.B.); (A.P.); (B.D.)
- Department of Chemical and Process Engineering, West Pomeranian University of Technology, Piastów 42, 71-311 Szczecin, Poland; (B.G.); (R.R.)
| | - Małgorzata Goszka
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (M.G.); (N.S.); (P.S.); (W.S.); (R.B.); (A.P.); (B.D.)
| | - Marta Gliźniewicz
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (M.G.); (N.S.); (P.S.); (W.S.); (R.B.); (A.P.); (B.D.)
| | - Bartłomiej Grygorcewicz
- Department of Chemical and Process Engineering, West Pomeranian University of Technology, Piastów 42, 71-311 Szczecin, Poland; (B.G.); (R.R.)
- Department of Forensic Genetic, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Natalia Serwin
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (M.G.); (N.S.); (P.S.); (W.S.); (R.B.); (A.P.); (B.D.)
| | - Patrycja Stodolak
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (M.G.); (N.S.); (P.S.); (W.S.); (R.B.); (A.P.); (B.D.)
| | - Weronika Słodzińska
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (M.G.); (N.S.); (P.S.); (W.S.); (R.B.); (A.P.); (B.D.)
| | - Radosław Birger
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (M.G.); (N.S.); (P.S.); (W.S.); (R.B.); (A.P.); (B.D.)
| | - Aleksandra Polikowska
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (M.G.); (N.S.); (P.S.); (W.S.); (R.B.); (A.P.); (B.D.)
| | - Marta Budkowska
- Department of Medical Analytics, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Rafał Rakoczy
- Department of Chemical and Process Engineering, West Pomeranian University of Technology, Piastów 42, 71-311 Szczecin, Poland; (B.G.); (R.R.)
| | - Barbara Dołęgowska
- Department of Laboratory Medicine, Pomeranian Medical University of Szczecin, Powstancow Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (M.G.); (N.S.); (P.S.); (W.S.); (R.B.); (A.P.); (B.D.)
| |
Collapse
|
27
|
Chen Y, Zhang Y, Jiang M, Ma H, Cai Y. HMOX1 as a therapeutic target associated with diabetic foot ulcers based on single-cell analysis and machine learning. Int Wound J 2024; 21:e14815. [PMID: 38468410 PMCID: PMC10928352 DOI: 10.1111/iwj.14815] [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: 12/23/2023] [Revised: 01/15/2024] [Accepted: 02/12/2024] [Indexed: 03/13/2024] Open
Abstract
Diabetic foot ulcers (DFUs) are a serious chronic complication of diabetes mellitus and a leading cause of disability and death in diabetic patients. However, current treatments remain unsatisfactory. Although macrophages are associated with DFU, their exact role in this disease remains uncertain. This study sought to detect macrophage-related genes in DFU and identify possible therapeutic targets. Single-cell datasets (GSE223964) and RNA-seq datasets (GSM68183, GSE80178, GSE134431 and GSE147890) associated with DFU were retrieved from the gene expression omnibus (GEO) database for this study. Analysis of the provided single-cell data revealed the distribution of macrophage subpopulations in the DFU. Four independent RNA-seq datasets were merged into a single DFU cohort and further analysed using bioinformatics. This included differential expression (DEG) analysis, multiple machine learning algorithms to identify biomarkers and enrichment analysis. Finally, key results were validated using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western bolt. Finally, the findings were validated using RT-qPCR and western blot. We obtained 802 macrophage-related genes in single-cell analysis. Differential expression analysis yielded 743 DEGs. Thirty-seven macrophage-associated DEGs were identified by cross-analysis of marker genes with macrophage-associated DEGs. Thirty-seven intersections were screened and cross-analysed using four machine learning algorithms. Finally, HMOX1 was identified as a potentially valuable biomarker. HMOX1 was significantly associated with biological pathways such as the insulin signalling pathway. The results showed that HMOX1 was significantly overexpressed in DFU samples. In conclusion, the analytical results of this study identified HMOX1 as a potentially valuable biomarker associated with macrophages in DFU. The results of our analysis improve our understanding of the mechanism of macrophage action in this disease and may be useful in developing targeted therapies for DFU.
Collapse
Affiliation(s)
- Yiqi Chen
- Department of Burn and Plastic SurgeryAffiliated Hospital of Nantong UniversityNantongChina
| | - Yixin Zhang
- Department of Burn and Plastic SurgeryAffiliated Hospital of Nantong UniversityNantongChina
- Department of Breast SurgeryYantai City Yantai Hill hospitalYantaiChina
| | - Ming Jiang
- Department of Burn and Plastic SurgeryAffiliated Hospital of Nantong UniversityNantongChina
| | - Hong Ma
- Department of Burn and Plastic SurgeryAffiliated Hospital of Nantong UniversityNantongChina
- Department of BurnHanzhong Central HospitalHanZhongChina
| | - Yuhui Cai
- Department of Burn and Plastic SurgeryAffiliated Hospital of Nantong UniversityNantongChina
| |
Collapse
|
28
|
Takizawa Y, Morino T, Takagi R, Otori N, Kojima H, Yamato M. Effect of basic fibroblast growth factor with collagen/gelatin fixture in a rabbit model of nasal septum perforation. Regen Ther 2024; 25:387-394. [PMID: 38425672 PMCID: PMC10901693 DOI: 10.1016/j.reth.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/21/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction The treatment of nasal septum perforation solely by surgical intervention presents significant challenges. This study evaluated the effect of basic fibroblast growth factor (bFGF) in combination with collagen/gelatin on wound healing of nasal septum perforation in a rabbit animal model. Methods A nasal septum perforation rabbit model was created. bFGF was added to a collagen/gelatin fixture and placed adjacent to the perforation, which is a complete defect. The rabbits were divided into three groups: the sham group that underwent the surgical procedure only, bFGF (-) group that received collagen/gelatin fixture without bFGF, and bFGF(+) group that received collagen/gelatin fixture with bFGF. The dimensions of the perforations were measured after 4 weeks, and the septum was subjected to histological examination. Results All perforations remained open in the sham group (closure rate: 20.4%-83.1%). The closure rates of the bFGF(-) and bFGF(+) groups were 49.4%-68.8% and 72.7%-100%, respectively. No significant difference was noted in the closure rates between the sham and bFGF(-) groups; however, significant differences were observed between the sham and bFGF(+) groups, and the bFGF(-) and bFGF(+) groups (p < 0.05), indicating that bFGF promoted perforation closure. Conclusions The study demonstrated that bFGF with collagen/gelatin carrier promoted wound healing in a rabbit model of nasal septum perforation.
Collapse
Affiliation(s)
- Yuki Takizawa
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
- Department of Otorhinolaryngology, Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, Japan
| | - Tsunetaro Morino
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
- Department of Otorhinolaryngology, Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, Japan
| | - Ryo Takagi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Nobuyoshi Otori
- Department of Otorhinolaryngology, Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, Japan
| | - Hiromi Kojima
- Department of Otorhinolaryngology, Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, Japan
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
29
|
Tejedor S, Wågberg M, Correia C, Åvall K, Hölttä M, Hultin L, Lerche M, Davies N, Bergenhem N, Snijder A, Marlow T, Dönnes P, Fritsche-Danielson R, Synnergren J, Jennbacken K, Hansson K. The Combination of Vascular Endothelial Growth Factor A (VEGF-A) and Fibroblast Growth Factor 1 (FGF1) Modified mRNA Improves Wound Healing in Diabetic Mice: An Ex Vivo and In Vivo Investigation. Cells 2024; 13:414. [PMID: 38474378 PMCID: PMC10930761 DOI: 10.3390/cells13050414] [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: 01/09/2024] [Revised: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Diabetic foot ulcers (DFU) pose a significant health risk in diabetic patients, with insufficient revascularization during wound healing being the primary cause. This study aimed to assess microvessel sprouting and wound healing capabilities using vascular endothelial growth factor (VEGF-A) and a modified fibroblast growth factor (FGF1). METHODS An ex vivo aortic ring rodent model and an in vivo wound healing model in diabetic mice were employed to evaluate the microvessel sprouting and wound healing capabilities of VEGF-A and a modified FGF1 both as monotherapies and in combination. RESULTS The combination of VEGF-A and FGF1 demonstrated increased vascular sprouting in the ex vivo mouse aortic ring model, and topical administration of a combination of VEGF-A and FGF1 mRNAs formulated in lipid nanoparticles (LNPs) in mouse skin wounds promoted faster wound closure and increased neovascularization seven days post-surgical wound creation. RNA-sequencing analysis of skin samples at day three post-wound creation revealed a strong transcriptional response of the wound healing process, with the combined treatment showing significant enrichment of genes linked to skin growth. CONCLUSION f-LNPs encapsulating VEGF-A and FGF1 mRNAs present a promising approach to improving the scarring process in DFU.
Collapse
Affiliation(s)
- Sandra Tejedor
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
- Systems Biology Research Center, School of Bioscience, University of Skövde, 541 28 Skövde, Sweden; (P.D.); (J.S.)
| | - Maria Wågberg
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
| | - Cláudia Correia
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
| | - Karin Åvall
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
| | - Mikko Hölttä
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
| | - Leif Hultin
- Imaging and Data Analytics, Clinical and Pharmacological Safety Science, BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden;
| | - Michael Lerche
- Advanced Drug Delivery, Pharmaceutical Science, BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden; (M.L.); (N.D.)
| | - Nigel Davies
- Advanced Drug Delivery, Pharmaceutical Science, BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden; (M.L.); (N.D.)
| | - Nils Bergenhem
- Alliance Management, Business Development and Licensing, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA 02451, USA
| | - Arjan Snijder
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden; (A.S.)
| | - Tom Marlow
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden; (A.S.)
| | - Pierre Dönnes
- Systems Biology Research Center, School of Bioscience, University of Skövde, 541 28 Skövde, Sweden; (P.D.); (J.S.)
- SciCross AB, 541 35 Skövde, Sweden
| | - Regina Fritsche-Danielson
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
| | - Jane Synnergren
- Systems Biology Research Center, School of Bioscience, University of Skövde, 541 28 Skövde, Sweden; (P.D.); (J.S.)
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Karin Jennbacken
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
| | - Kenny Hansson
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden (C.C.); (K.Å.); (K.J.)
| |
Collapse
|
30
|
Voza FA, Huerta CT, Le N, Shao H, Ribieras A, Ortiz Y, Atkinson C, Machuca T, Liu ZJ, Velazquez OC. Fibroblasts in Diabetic Foot Ulcers. Int J Mol Sci 2024; 25:2172. [PMID: 38396848 PMCID: PMC10889208 DOI: 10.3390/ijms25042172] [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: 12/15/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Fibroblasts are stromal cells ubiquitously distributed in the body of nearly every organ tissue. These cells were previously considered to be "passive cells", solely responsible for ensuring the turnover of the extracellular matrix (ECM). However, their versatility, including their ability to switch phenotypes in response to tissue injury and dynamic activity in the maintenance of tissue specific homeostasis and integrity have been recently revealed by the innovation of technological tools such as genetically modified mouse models and single cell analysis. These highly plastic and heterogeneous cells equipped with multifaceted functions including the regulation of angiogenesis, inflammation as well as their innate stemness characteristics, play a central role in the delicately regulated process of wound healing. Fibroblast dysregulation underlies many chronic conditions, including cardiovascular diseases, cancer, inflammatory diseases, and diabetes mellitus (DM), which represent the current major causes of morbidity and mortality worldwide. Diabetic foot ulcer (DFU), one of the most severe complications of DM affects 40 to 60 million people. Chronic non-healing DFU wounds expose patients to substantial sequelae including infections, gangrene, amputation, and death. A complete understanding of the pathophysiology of DFU and targeting pathways involved in the dysregulation of fibroblasts are required for the development of innovative new therapeutic treatments, critically needed for these patients.
Collapse
Affiliation(s)
- Francesca A. Voza
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
| | - Carlos Theodore Huerta
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
| | - Nga Le
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Hongwei Shao
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Antoine Ribieras
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
| | - Yulexi Ortiz
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Carl Atkinson
- Department of Internal Medicine, Division of Pulmonary Critical Care & Sleep Medicine, University of Florida, Gainesville, FL 32611, USA;
| | - Tiago Machuca
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
| | - Zhao-Jun Liu
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Omaida C. Velazquez
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
31
|
Chauhan S, Gulia M, Singh RP, Jhawat V. Diabetic Wound: Pathophysiology, Complications and Treatment Strategies. Curr Protein Pept Sci 2024; 25:200-205. [PMID: 37909438 DOI: 10.2174/0113892037276171231016103320] [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: 08/07/2023] [Revised: 09/26/2023] [Accepted: 10/03/2023] [Indexed: 11/03/2023]
Abstract
Diabetic wound healing is expected to affect 25% of all diabetics, resulting in less severe external factors, economic costs, and less trauma. Topical formulations have been continually improved to achieve a range of amazing properties and have had a significant impact on the management of diabetic wounds. Topical insulin has become one of the most attractive and convenient wound healing techniques due to its excellent biocompatibility, water retention, and therapeutic properties. Multiple versatile topical insulins have been identified and have shown promise over the past few years as they greatly facilitate the management of diabetic wounds as we understand their etiology. The physiological wound healing process repairs damaged tissue and restores skin integrity. For about a century, insulin, a powerful healing agent, and it has been utilized in several clinical and experimental researches research studies to accelerate the healing of various injuries.
Collapse
Affiliation(s)
- Sunita Chauhan
- Department of Pharmaceutical Science, School of Medical and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Monika Gulia
- Department of Pharmaceutical Science, School of Medical and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Rahul Pratap Singh
- Department of Pharmaceutical Science, School of Medical and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Vikas Jhawat
- Department of Pharmaceutical Science, School of Medical and Allied Science, GD Goenka University, Gurugram, Haryana, India
| |
Collapse
|
32
|
Zheng A, Xu Y, Cen N, Wu B. A Lower IL-34 Expression Is Associated with Non-Healing Diabetic Foot Ulcers. Comb Chem High Throughput Screen 2024; 27:1533-1543. [PMID: 37888825 DOI: 10.2174/0113862073273222231005065757] [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: 08/08/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND The non-healing of diabetic foot ulcers (DFU) is a major cause of high disability, morbidity, and mortality. Thus, new therapeutic targets and methods to help healing in patients with DFUs are major research hotspots. OBJECTIVE This study examined the molecular differences between healing and non-healing DFUs to identify genes associated with DFU healing. METHODS Differentially expressed genes (DEGs) were identified by bioinformatics. Samples were collected from patients with healing (n=10) and non-healing (n=10) DFUs from September 2021 to September 2022. Interleukin (IL)-34 expression was measured by ELISA and qRT-PCT. The fibroblasts from healing and non-healing DFU were divided according to their gene signatures and subdivided based on their gene expression profile differences. RESULTS A comparison of fibroblast subpopulation characteristics revealed that the proportion of subpopulation 4 was significantly higher in non-healing DFUs than in healing DFUs. Subpopulation 4 had 254 upregulated genes and 2402 downregulated genes in the non-healing compared with the healing DFUs. The DEGs were involved in several biological functions, including cytokine activity, receptor-ligand activity, signaling receptor activator activity, and receptor regulator activity. IL-34 was downregulated in non-healing compared with healing DFUs, suggesting a possible role of IL-34 in DFU healing. In the clinical specimens, IL-34 was significantly downregulated in non-healing DFUs, consistent with the bioinformatics results. CONCLUSION IL-34 expression is downregulated in non-healing DFU. IL-34 appears to be involved in DFU healing, but the exact causal relationship remains to be explored.
Collapse
Affiliation(s)
- Aitian Zheng
- Jinan University, Guangzhou, 511486, China
- Department of Endocrinology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Yuanyuan Xu
- Department of Endocrinology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Nimiao Cen
- Department of Endocrinology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Biaoliang Wu
- Jinan University, Guangzhou, 511486, China
- Department of Endocrinology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
| |
Collapse
|
33
|
Zhang H, Gu Y, Zhang K, Tu Y, Ouyang C. Roles and mechanisms of umbilical cord mesenchymal stem cells in the treatment of diabetic foot: A review of preclinical and clinical studies. J Diabetes Complications 2024; 38:108671. [PMID: 38154217 DOI: 10.1016/j.jdiacomp.2023.108671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/29/2023] [Accepted: 12/17/2023] [Indexed: 12/30/2023]
Abstract
AIMS Growing preclinical and clinical evidence has suggested the potential method of umbilical cord mesenchymal stem cell (UCMSC) therapy for diabetic foot. Thus, the authors provided an outline of the application of UCMSCs in the treatment of diabetic foot and further summarized the roles and mechanisms of this therapy. DATA SYNTHESIS With no time limitations, the authors searched the Web of Science, Cochrane Central Register of Controlled Trials, and PubMed (MEDLINE) databases. 14 studies were included, including 9 preclinical experiments and 5 clinical trials (3 RCTs and 2 single-arm trials). CONCLUSIONS The UCMSCs are of great efficacy and safety, and function mainly by reducing inflammation, regulating immunity, promoting growth factors, and enhancing the functions of vascular endothelial cells, fibroblasts, and keratinocytes. As a result, ulcer healing-related biological processes ensue, which finally lead to diabetic foot ulcer healing and clinical symptom improvement. UCMSC treatment enhances diabetic foot ulcer healing and has a safety profile. They function mainly by modulating immunity, promoting growth factor secretion, and enhancing cellular functions. More well-designed preclinical and clinical studies are needed to provide the most optimal protocol, the comprehensive molecular mechanisms, as well as to further evaluate the efficiency and safety profile of UCMSC treatment in diabetic foot patients.
Collapse
Affiliation(s)
- Haorui Zhang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing 100037, China
| | - Yuanrui Gu
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing 100037, China
| | - Ke Zhang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing 100037, China
| | - Yanxia Tu
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing 100037, China
| | - Chenxi Ouyang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing 100037, China.
| |
Collapse
|
34
|
Ye P, Gu R, Zhu H, Chen J, Han F, Nie X. SOX family transcription factors as therapeutic targets in wound healing: A comprehensive review. Int J Biol Macromol 2023; 253:127243. [PMID: 37806414 DOI: 10.1016/j.ijbiomac.2023.127243] [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: 09/07/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
The SOX family plays a vital role in determining the fate of cells and has garnered attention in the fields of cancer research and regenerative medicine. It also shows promise in the study of wound healing, as it actively participates in the healing processes of various tissues such as skin, fractures, tendons, and the cornea. However, our understanding of the mechanisms behind the SOX family's involvement in wound healing is limited compared to its role in cancer. Gaining insight into its role, distribution, interaction with other factors, and modifications in traumatized tissues could provide valuable new knowledge about wound healing. Based on current research, SOX2, SOX7, and SOX9 are the most promising members of the SOX family for future interventions in wound healing. SOX2 and SOX9 promote the renewal of cells, while SOX7 enhances the microvascular environment. The SOX family holds significant potential for advancing wound healing research. This article provides a comprehensive review of the latest research advancements and therapeutic tools related to the SOX family in wound healing, as well as the potential benefits and challenges of targeting the SOX family for wound treatment.
Collapse
Affiliation(s)
- Penghui Ye
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Rifang Gu
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; School Medical Office, Zunyi Medical University, Zunyi 563006, China
| | - Huan Zhu
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Jitao Chen
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Felicity Han
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xuqiang Nie
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
35
|
Chen L, Shen S, Wang S. LncRNA SNHG16 Knockdown Promotes Diabetic Foot Ulcer Wound Healing via Sponging MiR-31-5p. TOHOKU J EXP MED 2023; 261:283-289. [PMID: 37704416 DOI: 10.1620/tjem.2023.j078] [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: 09/15/2023]
Abstract
Diabetic foot ulcers are caused by nerve abnormalities and vascular lesions in the distal lower limbs of diabetic patients. However, the causes of diabetic foot ulcers are diverse and the treatment process is complex. Therefore, understanding the pathogenesis of diabetic foot ulcers through lncRNA and formulating effective means are the key to the cure of patients. Tissues were collected from 76 diabetic foot ulcer patients and 50 non-diabetic patients undergoing traumatic amputation. Human dermal fibroblasts (HDFs) were induced by high glucose to obtain diabetic foot ulcer cell model. The lncRNA SNHG16 (SNHG16) and miR-31-5p expression in tissues and cells was detected by real-time quantitative reverse transcription PCR (RT-qPCR). Cell Counting Kit-8 (CCK-8) and Transwell assays were used to evaluate the biological behavior of the cells, and the association between SNHG16 and miR-31-5p was explored by luciferase reporting assay. SNHG16 was distinctly expressed in diabetic foot ulcer tissue samples, while miR-31-5p was decreased. In vitro cell function assays confirmed that the proliferation level was inhibited in the constructed diabetic foot ulcer cell model (HG group), as was the migration and invasion ability. After transfection with silencing SNHG16, the biological behavior of the cells was promoted. Mechanistically, SNHG16 sponge miR-31-5p regulated disease progression. Recovery experiments revealed that miR-31-5p inhibitor counteracted the effect of silencing SNHG16 on cell viability. SNHG16 knockdown may regulate the biological function of cells by targeting miR-31-5p to promote wound healing and ameliorate the condition of diabetic foot ulcer patients.
Collapse
Affiliation(s)
- Lifen Chen
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University
| | - Shuyi Shen
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University
| | - Songyu Wang
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University
| |
Collapse
|
36
|
Toma AI, Shah D, Roth D, Oliver Piña J, Hymel L, Turner T, Kamalakar A, Liu K, Bartsch P, Jacobs L, D'Souza R, Liotta D, Botchwey E, Willett NJ, Goudy SL. Harnessing Bilayer Biomaterial Delivery of FTY720 as an Immunotherapy to Accelerate Oral Wound Healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.22.573096. [PMID: 38187740 PMCID: PMC10769397 DOI: 10.1101/2023.12.22.573096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Orofacial clefts are the most common craniofacial congenital anomaly. Following cleft palate repair, up to 60% of surgeries have wound healing complications leading to oronasal fistula (ONF), a persistent connection between the roof of the mouth and the nasal cavity. The current gold standard methods for ONF repair use human allograft tissues; however, these procedures have risks of graft infection and/or rejection, requiring surgical revisions. Immunoregenerative therapies present a novel alternative approach to harness the body's immune response and enhance the wound healing environment. We utilized a repurposed FDA-approved immunomodulatory drug, FTY720, to reduce the egress of lymphocytes and induce immune cell fate switching toward pro-regenerative phenotypes. Here, we engineered a bilayer biomaterial system using Tegaderm™, a liquid-impermeable wound dressing, to secure and control the delivery of FTY720- nanofiber scaffolds (FTY720-NF). We optimized release kinetics of the bilayer FTY720-NF to sustain drug release for up to 7d with safe, efficacious transdermal absorption and tissue biodistribution. Through comprehensive immunophenotyping, our results illustrate a pseudotime pro-regenerative state transition in recruited hybrid immune cells to the wound site. Additional histological assessments established a significant difference in full thickness ONF closure in mice on Day 7 following treatment with bilayer FTY720-NF, compared to controls. These findings demonstrate the utility of immunomodulatory strategies for oral wound healing, better positing the field to develop more efficacious treatment options for pediatric patients. One Sentence Summary Local delivery of bilayer FTY720-nanofiber scaffolds in an ONF mouse model promotes complete wound closure through modulation of pro-regenerative immune and stromal cells.
Collapse
|
37
|
Melamed E, Borkow G. Continuum of care in hard-to-heal wounds by copper dressings: a case series. J Wound Care 2023; 32:788-796. [PMID: 38060415 DOI: 10.12968/jowc.2023.32.12.788] [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: 12/18/2023]
Abstract
OBJECTIVE The quest for an ideal wound dressing has been a longstanding challenge due to the complex nature of wound healing, including stages of haemostasis, inflammation, maturation and remodelling, with overlapping timelines. This makes it difficult to find a single dressing that optimally supports all phases of wound healing. In addition, the ideal wound dressing should possess antibacterial properties and be capable of effectively debriding and lysing necrotic tissue. Copper is an essential trace element that participates in many of the key physiological wound healing processes. METHOD Copper stimulates secretion of various cytokines and growth factors, thus promoting angiogenesis, granulation tissue formation, extracellular matrix proteins secretion and re-epithelialisation. Harnessing this knowledge, we have used copper oxide-impregnated wound dressings in numerous cases and observed their benefits throughout the entire wound healing process. RESULTS This led us to postulate the 'continuum of care' hypothesis of copper dressings. In this study we describe four cases of hard-to-heal wounds of various aetiologies, in which we applied copper dressings consistently across all stages of wound healing, with rapid uneventful healing. CONCLUSION We believe we have successfully implemented the continuum of care principle.
Collapse
Affiliation(s)
- Eyal Melamed
- Foot and Ankle Service, Department of Orthopaedics, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Gadi Borkow
- The Skin Research Institute, The Dead-Sea & Arava Science Center, Masada 8693500, Israel
- MedCu Technologies Ltd., Herzliya 4672200, Israel
| |
Collapse
|
38
|
Dordoe C, Huang W, Bwalya C, Wang X, Shen B, Wang H, Wang J, Ye S, Wang P, Xiaoyan B, Li X, Lin L. The role of microglial activation on ischemic stroke: Modulation by fibroblast growth factors. Cytokine Growth Factor Rev 2023; 74:122-133. [PMID: 37573252 DOI: 10.1016/j.cytogfr.2023.07.005] [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: 07/21/2023] [Accepted: 07/29/2023] [Indexed: 08/14/2023]
Abstract
Stroke is one of the devastating clinical conditions that causes death and permanent disability. Its occurrence causes the reduction of oxygen and glucose supply, resulting in events such as inflammatory response, oxidative stress, and apoptosis in the brain. Microglia are brain-resident immune cells in the central nervous system (CNS) that exert diverse roles and respond to pathological process after an ischemic insult. The discovery of fibroblast growth factors (FGFs) in mammals, resulted to the findings that they can treat experimental models of stroke in animals effectively. FGFs function as homeostatic factors that control cells and hormones involved in metabolism, and they also regulate the secretion of proinflammatory (M1) and anti-inflammatory (M2) cytokines after stroke. In this review, we outline current evidence of microglia activation in experimental models of stroke focusing on its ability to exacerbate damage or repair tissue. Also, our review sheds light on the pharmacological actions of FGFs on multiple targets to regulate microglial modulation and highlighted their theoretical molecular mechanisms to provide possible therapeutic targets, as well as their limitations for the treatment of stroke. DATA AVAILABILITY: Not applicable.
Collapse
Affiliation(s)
- Confidence Dordoe
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wenting Huang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Canol Bwalya
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xue Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Bixin Shen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hao Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jing Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shasha Ye
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Peng Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Bao Xiaoyan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaokun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, Wenzhou, Zhejiang 325035, China.
| | - Li Lin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
39
|
Ma X, Zhang Y, Jiang J, Ru Y, Luo Y, Luo Y, Fei X, Song J, Ma X, Li B, Tan Y, Kuai L. Metabolism-related biomarkers, molecular classification, and immune infiltration in diabetic ulcers with validation. Int Wound J 2023; 20:3498-3513. [PMID: 37245869 PMCID: PMC10588317 DOI: 10.1111/iwj.14223] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/21/2023] [Indexed: 05/30/2023] Open
Abstract
Diabetes mellitus (DM) can lead to diabetic ulcers (DUs), which are the most severe complications. Due to the need for more accurate patient classifications and diagnostic models, treatment and management strategies for DU patients still need improvement. The difficulty of diabetic wound healing is caused closely related to biological metabolism and immune chemotaxis reaction dysfunction. Therefore, the purpose of our study is to identify metabolic biomarkers in patients with DU and construct a molecular subtype-specific prognostic model that is highly accurate and robust. RNA-sequencing data for DU samples were obtained from the Gene Expression Omnibus (GEO) database. DU patients and normal individuals were compared regarding the expression of metabolism-related genes (MRGs). Then, a novel diagnostic model based on MRGs was constructed with the random forest algorithm, and classification performance was evaluated utilizing receiver operating characteristic (ROC) analysis. The biological functions of MRGs-based subtypes were investigated using consensus clustering analysis. A principal component analysis (PCA) was conducted to determine whether MRGs could distinguish between subtypes. We also examined the correlation between MRGs and immune infiltration. Lastly, qRT-PCR was utilized to validate the expression of the hub MRGs with clinical validations and animal experimentations. Firstly, 8 metabolism-related hub genes were obtained by random forest algorithm, which could distinguish the DUs from normal samples validated by the ROC curves. Secondly, DU samples could be consensus clustered into three molecular classifications by MRGs, verified by PCA analysis. Thirdly, associations between MRGs and immune infiltration were confirmed, with LYN and Type 1 helper cell significantly positively correlated; RHOH and TGF-β family remarkably negatively correlated. Finally, clinical validations and animal experiments of DU skin tissue samples showed that the expressions of metabolic hub genes in the DU groups were considerably upregulated, including GLDC, GALNT6, RHOH, XDH, MMP12, KLK6, LYN, and CFB. The current study proposed an auxiliary MRGs-based DUs model while proposing MRGs-based molecular clustering and confirmed the association with immune infiltration, facilitating the diagnosis and management of DU patients and designing individualized treatment plans.
Collapse
Affiliation(s)
- Xiao‐Xuan Ma
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of DermatologyShanghai Academy of Traditional Chinese MedicineshanghaiChina
| | - Ying Zhang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of DermatologyShanghai Academy of Traditional Chinese MedicineshanghaiChina
| | - Jing‐Si Jiang
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Yi Ru
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of DermatologyShanghai Academy of Traditional Chinese MedicineshanghaiChina
| | - Ying Luo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of DermatologyShanghai Academy of Traditional Chinese MedicineshanghaiChina
| | - Yue Luo
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Xiao‐Ya Fei
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Jian‐Kun Song
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Xin Ma
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of DermatologyShanghai Academy of Traditional Chinese MedicineshanghaiChina
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Bin Li
- Institute of DermatologyShanghai Academy of Traditional Chinese MedicineshanghaiChina
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Yi‐Mei Tan
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Le Kuai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of DermatologyShanghai Academy of Traditional Chinese MedicineshanghaiChina
| |
Collapse
|
40
|
Sezen S, Ertuğrul MS, Balpınar Ö, Bayram C, Özkaraca M, Okkay IF, Hacımüftüoğlu A, Güllüce M. Assessment of antimicrobial activity and In Vitro wound healing potential of ZnO nanoparticles synthesized with Capparis spinosa extract. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:117609-117623. [PMID: 37872332 DOI: 10.1007/s11356-023-30417-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/08/2023] [Indexed: 10/25/2023]
Abstract
Agents that will accelerate wound healing maintain their clinical importance in all aspects. The aim of this study is to determine the antimicrobial activity of zinc oxide nanoparticles (ZnO NPs) ZnO nanoparticles obtained by green synthesis from Capparis spinosa L. extract and their effect on in vitro wound healing. ZnO NPs were synthesized and characterized using Capparis spinosa L. extract. ZnO NPs were tested against nine ATCC-coded pathogen strains to determine antimicrobial activity. The effects of different doses (0.0390625-20 µg/mL) of NPs on cell viability were determined by MTT assay. The effect of ZnO NPs doses (0.0390625 µg/mL, 0.078125 µg/mL, 0.15625 µg/mL, 0.3125 µg/mL, 0.625 µg/mL, 1.25 µg/mL) that increase proliferation and migration on wound healing was investigated in an in vitro wound experiment. Cell culture medium obtained from the in vitro wound assay was used for biochemical analysis, and plate alcohol-fixed cells were used for immunohistochemical staining. It was determined that NPs formed an inhibition zone against the tested Gram-positive bacteria. The ZnO NPs doses determined in the MTT test provided faster wound closure in in-vitro conditions compared to the DMSO group. Biochemical analyses showed that inflammation and oxidative status decreased, while antioxidant levels increased in ZnO NPs groups. Immunohistochemical analyses showed increased expression levels of Bek/FGFR2, IGF, and TGF-β associated with wound healing. The findings reveal the antimicrobial effect of ZnO nanoparticles obtained using Capparis spinosa L. extract in vitro and their potential applications in wound healing.
Collapse
Affiliation(s)
- Selma Sezen
- Department of Medical Pharmacology, Faculty of Medicine, Agri Ibrahim Cecen University, Agri, Türkiye
| | | | - Özge Balpınar
- Hemp Research Institute, Ondokuz Mayıs University, Samsun, Türkiye
| | - Cemil Bayram
- Department of Pharmacology and Toxicology, Faculty of Veterinary, Ataturk University, Erzurum, Türkiye
| | - Mustafa Özkaraca
- Department of Pathology, Faculty of Veterinary, Sivas Cumhuriyet University, Sivas, Türkiye
| | - Irmak Ferah Okkay
- Department of Pharmacology, Faculty of Pharmacy, Ataturk University, Erzurum, Türkiye
| | - Ahmet Hacımüftüoğlu
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Türkiye
| | - Medine Güllüce
- Department of Biology, Faculty of Science, Ataturk University, Erzurum, Türkiye
| |
Collapse
|
41
|
Li H, Jing S, Xu H. Effect and mechanism of microRNAs on various diabetic wound local cells. J Diabetes 2023; 15:955-967. [PMID: 37679063 PMCID: PMC10667630 DOI: 10.1111/1753-0407.13474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/20/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
The difficulty of wound healing in diabetes mellitus has long been regarded as a thorny problem in the medical field. One of the important reasons is the abnormal function of wound-related cells. A large number of recent studies have shown that microRNA (miR), a noncoding RNA that exists in eukaryotic cells, is closely linked to the functions of various cells in diabetic wound, and ultimately affects the healing of wound. This paper establishes for the first time the connection between miR and wound healing from the cellular perspective and summarizes the effects of various miRs on one or more kinds of wound cells, including their targets and related mechanisms. The abnormal expression of miRs in the wound has certain value for the early diagnosis of diabetic wounds. Moreover, it seems that correcting miRs that are abnormal expressed in the wound or artificially adding miRs that can promote wound healing has an essential therapeutic value.
Collapse
Affiliation(s)
- Hongjie Li
- Department of Vascular SurgeryThe Third Xiangya Hospital of Central South UniversityChangshaChina
- Central South University Xiangya School of MedicineChangshaChina
| | - Shengyu Jing
- Department of Vascular SurgeryThe Third Xiangya Hospital of Central South UniversityChangshaChina
- Central South University Xiangya School of MedicineChangshaChina
| | - Hongbo Xu
- Department of Vascular SurgeryThe Third Xiangya Hospital of Central South UniversityChangshaChina
| |
Collapse
|
42
|
Zhang W, He Y, Zhang Y. CircRNA in ocular neovascular diseases: Fundamental mechanism and clinical potential. Pharmacol Res 2023; 197:106946. [PMID: 37797661 DOI: 10.1016/j.phrs.2023.106946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023]
Abstract
Ocular neovascular disease (OND), characterized by the aberrant formation of immature blood vessels, is the leading cause of vision impairment and blindness. It is important to find effective ways to diagnose and treat these diseases. Circular RNA (circRNA) is a group of endogenous non-coding RNA that play a crucial role in regulating different biological processes. Due to their close association with ocular disease and angiogenesis, circRNAs have become a hotspot in OND research. In this review, we intensively investigate the possibility of using circRNAs in the management of ONDs. In general, angiogenesis is divided into five phases. On the basis of these five steps, we describe the potential of using circRNAs by introducing how they regulate angiogenesis. Subsequently, the interactions between circRNAs and ONDs, including pterygium, corneal neovascularization, age-related macular degeneration, diabetic retinopathy, and retinopathy of prematurity, are analyzed in detail. We also introduce the potential use of circRNAs as OND diagnostic biomarkers. Finally, we summarize the prospects of using circRNAs as a potential strategy in OND management. The gaps in recent research are also pointed out with the purpose of promoting the introduction of circRNAs into clinical applications.
Collapse
Affiliation(s)
- Wenxin Zhang
- Department of Ophthalmology, 2nd Hospital of Jilin University, Changchun 130041, Jilin, China
| | - Yuxi He
- Department of Ophthalmology, 2nd Hospital of Jilin University, Changchun 130041, Jilin, China
| | - Yan Zhang
- Department of Ophthalmology, 2nd Hospital of Jilin University, Changchun 130041, Jilin, China; Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| |
Collapse
|
43
|
Rai V, Le H, Agrawal DK. Novel mediators regulating angiogenesis in diabetic foot ulcer healing. Can J Physiol Pharmacol 2023; 101:488-501. [PMID: 37459652 DOI: 10.1139/cjpp-2023-0193] [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: 09/08/2023]
Abstract
A non-healing diabetic foot ulcer (DFU) is a debilitating clinical problem amounting to socioeconomic and psychosocial burdens. DFUs increase morbidity due to prolonged treatment and mortality in the case of non-treatable ulcers resulting in gangrene and septicemia. The overall amputation rate of the lower extremity with DFU ranges from 3.34% to 42.83%. Wound debridement, antibiotics, applying growth factors, negative pressure wound therapy, hyperbaric oxygen therapy, topical oxygen, and skin grafts are common therapies for DFU. However, recurrence and nonhealing ulcers are still major issues. Chronicity of inflammation, hypoxic environment, poor angiogenesis, and decreased formation of the extracellular matrix (ECM) are common impediments leading to nonhealing patterns of DFUs. Angiogenesis is crucial for wound healing since proper vessel formation facilitates nutrients, oxygen, and immune cells to the ulcer tissue to help in clearing out debris and facilitate healing. However, poor angiogenesis due to decreased expression of angiogenic mediators and matrix formation results in nonhealing and ultimately amputation. Multiple proangiogenic mediators and vascular endothelial growth factor (VEGF) therapy exist to enhance angiogenesis, but the results are not satisfactory. Thus, there is a need to investigate novel pro-angiogenic mediators that can either alone or in combination enhance the angiogenesis and healing of DFUs. In this article, we critically reviewed the existing pro-angiogenic mediators followed by potentially novel factors that might play a regulatory role in promoting angiogenesis and wound healing in DFUs.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Hoangvi Le
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
44
|
Jere SW, Abrahamse H, Houreld NN. Interaction of the AKT and β-catenin signalling pathways and the influence of photobiomodulation on cellular signalling proteins in diabetic wound healing. J Biomed Sci 2023; 30:81. [PMID: 37735655 PMCID: PMC10515080 DOI: 10.1186/s12929-023-00974-8] [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: 06/26/2023] [Accepted: 09/14/2023] [Indexed: 09/23/2023] Open
Abstract
The induction of a cells destiny is a tightly controlled process that is regulated through communication between the matrix and cell signalling proteins. Cell signalling activates distinctive subsections of target genes, and different signalling pathways may be used repeatedly in different settings. A range of different signalling pathways are activated during the wound healing process, and dysregulated cellular signalling may lead to reduced cell function and the development of chronic wounds. Diabetic wounds are chronic and are characterised by the inability of skin cells to act in response to reparative inducements. Serine/threonine kinase, protein kinase B or AKT (PKB/AKT), is a central connection in cell signalling induced by growth factors, cytokines and other cellular inducements, and is one of the critical pathways that regulate cellular proliferation, survival, and quiescence. AKT interacts with a variety of other pathway proteins including glycogen synthase kinase 3 beta (GSK3β) and β-catenin. Novel methodologies based on comprehensive knowledge of activated signalling pathways and their interaction during normal or chronic wound healing can facilitate quicker and efficient diabetic wound healing. In this review, we focus on interaction of the AKT and β-catenin signalling pathways and the influence of photobiomodulation on cellular signalling proteins in diabetic wound healing.
Collapse
Affiliation(s)
- Sandy Winfield Jere
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein, 2028 South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein, 2028 South Africa
| | - Nicolette Nadene Houreld
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein, 2028 South Africa
| |
Collapse
|
45
|
Li X, Jing X, Yu Z, Huang Y. Diverse Antibacterial Treatments beyond Antibiotics for Diabetic Foot Ulcer Therapy. Adv Healthc Mater 2023; 12:e2300375. [PMID: 37141030 DOI: 10.1002/adhm.202300375] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/06/2023] [Indexed: 05/05/2023]
Abstract
Diabetic foot ulcer (DFU), a common complication of diabetes, has become a great burden to both patients and the society. The delayed wound closure of ulcer sites resulting from vascular damage and neutrophil dysfunction facilitates bacterial infection. Once drug resistance occurs or bacterial biofilm is formed, conventional therapy tends to fail and amputation is unavoidable. Therefore, effective antibacterial treatment beyond antibiotics is of utmost importance to accelerate the wound healing process and prevent amputation. Considering the complexity of multidrug resistance, biofilm formation, and special microenvironments (such as hyperglycemia, hypoxia, and abnormal pH value) at the infected site of DFU, several antibacterial agents and different mechanisms have been explored to achieve the desired outcome. The present review focuses on the recent progress of antibacterial treatments, including metal-based medications, natural and synthesized antimicrobial peptides, antibacterial polymers, and sensitizer-based therapy. This review provides a valuable reference for the innovation of antibacterial material design for DFU therapy.
Collapse
Affiliation(s)
- Xiaoyuan Li
- Faculty of Chemistry, Northeast Normal University, Renmin Street, Changchun, 130024, P. R. China
| | - Xin Jing
- Faculty of Chemistry, Northeast Normal University, Renmin Street, Changchun, 130024, P. R. China
| | - Ziqian Yu
- Faculty of Chemistry, Northeast Normal University, Renmin Street, Changchun, 130024, P. R. China
| | - Yubin Huang
- Faculty of Chemistry, Northeast Normal University, Renmin Street, Changchun, 130024, P. R. China
| |
Collapse
|
46
|
Ortega-Sánchez C, Pérez-Díaz M, Melgarejo-Ramírez Y, Chopin-Doroteo M, Silva-Bermudez P, Martínez-López V, Zacaula-Juárez N, Zamudio-Cuevas Y, Hernández-Valencia C, López-Jácome LE, Carlos-Martínez A, Reyes-Medina N, Tamez-Pedroza L, Martínez-Pardo ME, Reyes-Frías MDL, Lecona H, Baeza I, Martinez-Gutierrez F, Márquez-Gutiérrez E, Martínez-Castañon G, Sánchez-Sánchez R. Radiosterilized Pig Skin, Silver Nanoparticles and Skin Cells as an Integral Dressing Treatment for Burns: Development, Pre-Clinical and Clinical Pilot Study. Pharmaceutics 2023; 15:2105. [PMID: 37631319 PMCID: PMC10458621 DOI: 10.3390/pharmaceutics15082105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Radiosterilized pig skin (RPS) has been used as a dressing for burns since the 1980s. Its similarity to human skin in terms of the extracellular matrix (ECM) allows the attachment of mesenchymal stem cells, making it ideal as a scaffold to create cellularized constructs. The use of silver nanoparticles (AgNPs) has been proven to be an appropriate alternative to the use of antibiotics and a potential solution against multidrug-resistant bacteria. RPS can be impregnated with AgNPs to develop nanomaterials capable of preventing wound infections. The main goal of this study was to assess the use of RPS as a scaffold for autologous fibroblasts (Fb), keratinocytes (Kc), and mesenchymal stem cells (MSC) in the treatment of second-degree burns (SDB). Additionally, independent RPS samples were impregnated with AgNPs to enhance their properties and further develop an antibacterial dressing that was initially tested using a burn mouse model. This protocol was approved by the Research and Ethics Committee of the INRLGII (INR 20/19 AC). Transmission electron microscopy (TEM) and dynamic light scattering (DLS) analysis of the synthesized AgNPs showed an average size of 10 nm and rounded morphology. Minimum inhibitory concentrations (MIC) and Kirby-Bauer assays indicated that AgNPs (in solution at a concentration of 125 ppm) exhibit antimicrobial activity against the planktonic form of S. aureus isolated from burned patients; moreover, a log reduction of 1.74 ± 0.24 was achieved against biofilm formation. The nanomaterial developed with RPS impregnated with AgNPs solution at 125 ppm (RPS-AgNPs125) facilitated wound healing in a burn mouse model and enhanced extracellular matrix (ECM) deposition, as analyzed by Masson's staining in histological samples. No silver was detected by energy-dispersive X-ray spectroscopy (EDS) in the skin, and neither by Inductively Coupled Plasma Mass Spectrometry (ICP-MS) in different organs of the mouse burn model. Calcein/ethidium homodimer (EthD-1), 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT), and scanning electron microscopy (SEM) analysis demonstrated that Fb, Kc, and MSC could attach to RPS with over 95% cell viability. Kc were capable of releasing FGF at 0.5 pg above control levels, as analyzed by ELISA assays. An autologous RPS-Fb-Kc construct was implanted in a patient with SDB and compared to an autologous skin graft. The patient recovery was assessed seven days post-implantation, and the patient was followed up at one, two, and three months after the implantation, exhibiting favorable recovery compared to the gold standard, as measured by the cutometer. In conclusion, RPS effectively can be used as a scaffold for the culture of Fb, Kc, and MSC, facilitating the development of a cellularized construct that enhances wound healing in burn patients.
Collapse
Affiliation(s)
- Carmina Ortega-Sánchez
- Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico; (C.O.-S.); (M.P.-D.); (Y.M.-R.); (N.Z.-J.)
| | - Mario Pérez-Díaz
- Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico; (C.O.-S.); (M.P.-D.); (Y.M.-R.); (N.Z.-J.)
- Laboratorio de Biomembranas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico;
| | - Yaaziel Melgarejo-Ramírez
- Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico; (C.O.-S.); (M.P.-D.); (Y.M.-R.); (N.Z.-J.)
| | - Mario Chopin-Doroteo
- Laboratorio de Tejido Conjuntivo, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico;
| | - Phaedra Silva-Bermudez
- Unidad de Ingeniería de Tejidos Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico; (P.S.-B.); (V.M.-L.)
| | - Valentín Martínez-López
- Unidad de Ingeniería de Tejidos Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico; (P.S.-B.); (V.M.-L.)
| | - Noé Zacaula-Juárez
- Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico; (C.O.-S.); (M.P.-D.); (Y.M.-R.); (N.Z.-J.)
| | - Yessica Zamudio-Cuevas
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico;
| | - Carmen Hernández-Valencia
- Departamento de Alimentos y Biotecnología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Luis Esaú López-Jácome
- Laboratorio de Infectología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico;
| | - Alberto Carlos-Martínez
- Laboratorio de Microscopía Electrónica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico; (A.C.-M.); (N.R.-M.)
| | - Naxieli Reyes-Medina
- Laboratorio de Microscopía Electrónica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico; (A.C.-M.); (N.R.-M.)
| | - Luis Tamez-Pedroza
- Cirugía Plástica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico;
| | - María Esther Martínez-Pardo
- Banco de Tejidos Radioesterilizados, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52045, Mexico; (M.E.M.-P.); (M.d.L.R.-F.)
| | - María de Lourdes Reyes-Frías
- Banco de Tejidos Radioesterilizados, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac 52045, Mexico; (M.E.M.-P.); (M.d.L.R.-F.)
| | - Hugo Lecona
- Bioterio, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico;
| | - Isabel Baeza
- Laboratorio de Biomembranas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico;
| | - Fidel Martinez-Gutierrez
- Laboratorio de Antimicrobianos, Biopelículas y Microbiota, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, Mexico;
- Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autonoma de San Luis Potosi, San Luis Potosi 78210, Mexico
| | - Erik Márquez-Gutiérrez
- Cirugía Plástica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico;
| | | | - Roberto Sánchez-Sánchez
- Unidad de Ingeniería de Tejidos Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico; (P.S.-B.); (V.M.-L.)
- Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Mexico City 64849, Mexico
| |
Collapse
|
47
|
Qin B, Wu S, Dong H, Deng S, Liu Y, Zhang W, Feng G, Lei L, Xie H. Accelerated Healing of Infected Diabetic Wounds by a Dual-Layered Adhesive Film Cored with Microsphere-Loaded Hydrogel Composite Dressing. ACS APPLIED MATERIALS & INTERFACES 2023; 15:33207-33222. [PMID: 37418597 DOI: 10.1021/acsami.2c22650] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/09/2023]
Abstract
Diabetic wounds, a prevalent chronic disease, are associated with older age. The hyperglycemic microenvironment in diabetic wounds significantly reduces the immune system, inducing bacterial invasion. The coupling of tissue repair and antibacterial treatment is critical for infected diabetic ulcer regeneration. In this study, a dual-layered sodium alginate/carboxymethyl chitosan (SA/CMCS) adhesive film cored with an SA-bFGF microsphere-loaded small intestine submucosa (SIS) hydrogel composite dressing with a graphene oxide (GO)-based antisense transformation system was developed to promote infected diabetic wound healing and bacterial eradication. Initially, our injectable SIS-based hydrogel composite stimulated angiogenesis, collagen deposition, and immunoregulation in diabetic wound repair. The GO-based transformation system subsequently inhibited bacterial viability in infected wounds by post-transformation regulation. Meanwhile, the SA/CMCS film provided stable adhesion covering the wound area to maintain a moist microenvironment, which promoted in situ tissue repair. Our findings provide a promising clinical translation strategy for promoting the healing of infected diabetic wounds.
Collapse
Affiliation(s)
- Boquan Qin
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Shizhou Wu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Hongxian Dong
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Shu Deng
- Boston University Henry M. Goldman School of Dental Medicine, Boston, Massachusetts 02215-1300, United States
| | - Yunjie Liu
- West China School of Public Health, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Wanli Zhang
- Core Facilities of West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Guoying Feng
- College of Electronics and Information Engineering, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lei Lei
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Huiqi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| |
Collapse
|
48
|
Garabet W, Shabes P, Wolters KH, Rembe JD, Ibing W, Wagenhäuser MU, Simon F, Schelzig H, Oberhuber A. Effect of Gelatin-Based Hemostats on Fibroblasts and Relevant Growth Factors in Wound Healing. Gels 2023; 9:504. [PMID: 37367174 DOI: 10.3390/gels9060504] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 06/28/2023] Open
Abstract
Gelatin-based hemostats have been used in various surgical fields and showed advantageous effects on central aspects of wound healing when compared to cellulose-based hemostats. Nevertheless, the influence of gelatin-based hemostats on wound healing has not been fully explored yet. Hemostats were applied to fibroblast cell cultures for 5, 30, 60 min, 24 h, 7 and 14 days and measurements were taken at 3, 6, 12, 24 h and 7 or 14 days, respectively. Cell proliferation was quantified after different exposure times and a contraction assay was conducted to measure the extent of the extracellular matrix over time. We further assessed quantitative levels of vascular endothelial growth factor and basic fibroblast growth factor using enzyme-linked immunosorbent assay. Fibroblast counts decreased significantly at 7 and 14 days independent of the application duration (p < 0.001 for 5 min application). The gelatin-based hemostat did not have a negative impact on cell matrix contraction. After application of gelatin-based hemostat, the basic fibroblast growth factor did not change; yet, the vascular endothelial growth factor significantly increased after a prolonged 24 h application time when compared to controls or to a 6 h exposure (p < 0.05). Gelatin-based hemostats did not impair contraction of the extracellular matrix or growth factor production (vascular endothelial growth factor and basic fibroblast growth factor), while cell proliferation diminished at late time points. In conclusion, the gelatin-based material seems to be compatible with central aspects of wound healing. For further clinical assessment, future animal and human studies are necessary.
Collapse
Affiliation(s)
- Waseem Garabet
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Polina Shabes
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Katharina Henrika Wolters
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Julian-Dario Rembe
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Wiebke Ibing
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Markus Udo Wagenhäuser
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Florian Simon
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Hubert Schelzig
- Department of Vascular and Endovascular Surgery, University Hospital of Düsseldorf, 40225 Düsseldorf, Germany
| | - Alexander Oberhuber
- Department of Vascular and Endovascular Surgery, University Hospital of Münster, 48149 Münster, Germany
| |
Collapse
|
49
|
Beheshtizadeh N, Salimi A, Golmohammadi M, Ansari JM, Azami M. In-silico engineering of RNA nanoplatforms to promote the diabetic wound healing. BMC Chem 2023; 17:52. [PMID: 37291669 PMCID: PMC10251717 DOI: 10.1186/s13065-023-00969-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Abstract
One of the most notable required features of wound healing is the enhancement of angiogenesis, which aids in the acceleration of regeneration. Poor angiogenesis during diabetic wound healing is linked to a shortage of pro-angiogenic or an increase in anti-angiogenic factors. As a result, a potential treatment method is to increase angiogenesis promoters and decrease suppressors. Incorporating microRNAs (miRNAs) and small interfering RNAs (siRNAs), two forms of quite small RNA molecules, is one way to make use of RNA interference. Several different types of antagomirs and siRNAs are now in the works to counteract the negative effects of miRNAs. The purpose of this research is to locate novel antagonists for miRNAs and siRNAs that target multiple genes to promote angiogenesis and wound healing in diabetic ulcers.In this context, we used gene ontology analysis by exploring across several datasets. Following data analysis, it was processed using a systems biology approach. The feasibility of incorporating the proposed siRNAs and miRNA antagomirs into polymeric bioresponsive nanocarriers for wound delivery was further investigated by means of a molecular dynamics (MD) simulation study. Among the three nanocarriers tested (Poly (lactic-co-glycolic acid) (PLGA), Polyethylenimine (PEI), and Chitosan (CTS), MD simulations show that the integration of PLGA/hsa-mir-422a is the most stable (total energy = -1202.62 KJ/mol, Gyration radius = 2.154 nm, and solvent-accessible surface area = 408.416 nm2). With values of -25.437 KJ/mol, 0.047 nm for the Gyration radius, and 204.563 nm2 for the SASA, the integration of the second siRNA/ Chitosan took the last place. The results of the systems biology and MD simulations show that the suggested RNA may be delivered through bioresponsive nanocarriers to speed up wound healing by boosting angiogenesis.
Collapse
Affiliation(s)
- Nima Beheshtizadeh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Students? Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Alireza Salimi
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Science, Bojnurd, Iran
| | - Mahsa Golmohammadi
- Department of Polymer Engineering and Color Technology, Amirkabir University of Technology, Tehran, Iran
| | - Javad Mohajer Ansari
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Anatomy, School of Medicine, Hormozgan University of Medical Sciences, Jomhuri Eslami Blvd, Bandar Abbas, 7919915519, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
50
|
Chang C, Guo W, Yu X, Guo C, Zhou N, Guo X, Huang RL, Li Q, Zhu Y. Engineered M13 phage as a novel therapeutic bionanomaterial for clinical applications: From tissue regeneration to cancer therapy. Mater Today Bio 2023; 20:100612. [PMID: 37063776 PMCID: PMC10102448 DOI: 10.1016/j.mtbio.2023.100612] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/12/2023] [Accepted: 03/15/2023] [Indexed: 04/03/2023] Open
Abstract
Bacteriophages (phages) are nanostructured viruses with highly selective antibacterial properties that have gained attention beyond eliminating bacteria. Specifically, M13 phages are filamentous phages that have recently been studied in various aspects of nanomedicine due to their biological advantages and more compliant engineering capabilities over other phages. Having nanofiber-like morphology, M13 phages can reach varied target sites and self-assemble into multidimensional scaffolds in a relatively safe and stable way. In addition, genetic modification of the coat proteins enables specific display of peptides and antibodies on the phages, allowing for precise and individualized medicine. M13 phages have also been subjected to novel engineering approaches, including phage-based bionanomaterial engineering and phage-directed nanomaterial combinations that enhance the bionanomaterial properties of M13 phages. In view of these features, researchers have been able to utilize M13 phages for therapeutic applications such as drug delivery, biodetection, tissue regeneration, and targeted cancer therapy. In particular, M13 phages have been utilized as a novel bionanomaterial for precisely mimicking natural tissue environment in order to overcome the shortage in tissue and organ donors. Hence, in this review, we address the recent studies and advances of using M13 phages in the field of nanomedicine as therapeutic agents based upon their characteristics as novel bionanomaterial with biomolecules displayed. This paper also emphasizes the novel engineering approach that enhances M13 phage's bionanomaterial capabilities. Current limitations and future approaches are also discussed to provide insight in further progress for M13 phage-based clinical applications.
Collapse
Affiliation(s)
- Cheng Chang
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Wennan Guo
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Xinbo Yu
- Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China
| | - Chaoyi Guo
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Nan Zhou
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Xiaokui Guo
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
| | - Ru-Lin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Corresponding author.
| | - Qingtian Li
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Corresponding author.
| | - Yongzhang Zhu
- School of Global Health, Chinese Centre for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, 200025, China
- Corresponding author.
| |
Collapse
|