1
|
Ruan J, Xia Y, Ma Y, Xu X, Luo S, Yi J, Wu B, Chen R, Wang H, Yu H, Yang Q, Wu W, Sun D, Zhong J. Milk-derived exosomes as functional nanocarriers in wound healing: Mechanisms, applications, and future directions. Mater Today Bio 2025; 32:101715. [PMID: 40242483 PMCID: PMC12003018 DOI: 10.1016/j.mtbio.2025.101715] [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: 02/06/2025] [Revised: 03/22/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Wound healing presents a significant challenge in healthcare, imposing substantial physiological and economic burdens. While traditional treatments and stem cell therapies have shown benefits, milk-derived exosomes (MDEs) offer distinct advantages as a cell-free therapeutic approach. MDEs, isolated from mammalian milk, are characterized by their biocompatibility, ease of acquisition, and high yield, making them a promising tool for enhancing wound repair. This review provides a comprehensive analysis of the composition, sources, and extraction methods of MDEs, with a focus on their therapeutic role in both acute and diabetic chronic wounds. MDEs facilitate wound healing through the delivery of bioactive molecules, modulating key processes such as inflammation, angiogenesis, and collagen synthesis. Their ability to regulate complex wound-healing pathways underscores their potential for widespread clinical application. This review highlights the importance of MDEs in advancing wound management and proposes strategies to optimize their use in regenerative medicine.
Collapse
Affiliation(s)
- Jing Ruan
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Yuping Xia
- Department of Burn and Plastic Surgery, Zigong Fourth People's Hospital, Zigong 643099, China
| | - Yilei Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Xiyao Xu
- Department of Burn and Plastic Surgery, Zigong Fourth People's Hospital, Zigong 643099, China
| | - Shihao Luo
- Department of Burn and Plastic Surgery, Zigong Fourth People's Hospital, Zigong 643099, China
| | - Jia Yi
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Baihui Wu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, 999077, Hong Kong Special Administrative Region of China
| | - Hanbing Wang
- Department of Biotechnology, The University of Hong Kong, 999077, Hong Kong Special Administrative Region of China
| | - Honggang Yu
- Hand and Foot Surgery, The Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu 322000, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing 400044, China
- Jin Feng Laboratory, Chongqing, 401329, China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Junbo Zhong
- Department of Burn and Plastic Surgery, Zigong Fourth People's Hospital, Zigong 643099, China
| |
Collapse
|
2
|
Choi YW, Han SK, Jeong SH, Dhong ES. Creating a Practical Tool for Predicting Major Amputation Rate in Patients With Diabetic Hindfoot Ulcers: Focus on Ischemia and Infection. J Korean Med Sci 2025; 40:e55. [PMID: 40329787 DOI: 10.3346/jkms.2025.40.e55] [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: 08/04/2024] [Accepted: 11/07/2024] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Patients with diabetic hindfoot ulcers typically harbor significant concerns regarding their prospects for healing or the potential for major amputation. Nonetheless, a scarcity of data addressing this prevalent and critical query exists. Thus, the aim of this study was to create an initial risk-scoring system to forecast the prognosis of individuals with diabetic hindfoot ulcers, leveraging assessments of ischemia and infection severity, which are recognized as the principal risk factors for amputation. METHODS Ischemia severity was categorized as iS0, iS1, or iS2 based on transcutaneous partial oxygen tension values, while infection severity was classified as iN0, iN1, or iN2 according to the results of tissue and bone biopsy cultures. Risk scores were determined by summing the scores for ischemia and infection severity, yielding a range of 0 to 4. Wound healing outcomes were graded as either healed with or without major amputation. Wound healing outcomes were assessed based on the assigned risk scores. RESULTS With ascending risk scores, the proportion of patients subjected to major amputation also increased (P value for trend < 0.001). Univariable logistic regression analysis revealed a significant positive correlation between escalating risk scores and major amputation incidence. Patients with a risk score of 4 exhibited a 41-fold higher likelihood of undergoing major amputation compared to those with a risk score of 0. CONCLUSION Risk scores can serve as a reliable predictor of the major amputation rate in patients with diabetic hindfoot ulcers.
Collapse
Affiliation(s)
- Ye-Won Choi
- Diabetic Wound Center and Department of Plastic Surgery, Korea University Guro Hospital, Seoul, Korea
| | - Seung-Kyu Han
- Diabetic Wound Center and Department of Plastic Surgery, Korea University Guro Hospital, Seoul, Korea
- Department of Plastic Surgery, Korea University College of Medicine, Seoul, Korea.
| | - Seong-Ho Jeong
- Department of Plastic Surgery, Korea University College of Medicine, Seoul, Korea
| | - Eun-Sang Dhong
- Department of Plastic Surgery, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
3
|
Kwon M, Lee Y, Kim KS. Biomimetic gradient hydrogel with fibroblast spheroids for full-thickness skin regeneration. BIOMATERIALS ADVANCES 2025; 169:214152. [PMID: 39708659 DOI: 10.1016/j.bioadv.2024.214152] [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: 07/21/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
Hydrogel-based scaffolds have been widely investigated for their use in tissue engineering to accelerate tissue regeneration. However, replicating the physiological microenvironments of tissues with appropriate biological cues remains challenging. Recent advances in gradient hydrogels have transformed tissue-engineering research by providing precise structures that mimic the extracellular matrix of natural tissues. Unlike conventional homogeneously structured hydrogels, gradient hydrogels provide a better bio-mimicking microenvironment for combined cell therapies in chronic wound treatment by regulating various cell behaviors, such as proliferation, migration, and differentiation. Here, we present the integration of L929 mouse fibroblast spheroids into gradient hydrogels to mimic the dermal stiffness microenvironment and we investigated their impact on full-thickness skin regeneration. A stiffness gradient was achieved by modulating the concentration of methacrylated hyaluronic acid (HA-MA) with varying degrees of methacrylation, using a dual-syringe pump system. The encapsulation of L929 spheroids with gradient hydrogel facilitated skin cell organization in a hierarchically ordered configuration, leading to full-thickness wound healing that was 1.53 times faster than the untreated group in a rat model. This study provides a method for investigating the potential role of gradient hydrogels in various tissue engineering and regeneration applications.
Collapse
Affiliation(s)
- Mina Kwon
- School of Chemical Engineering, Pusan National University, Busan 46241, Republic of Korea.
| | - Yuhan Lee
- Department of Anaesthesiology, Perioperative and Pain Medicine, Center for Accelerated Medical Innovation, Center for Nanomedicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America.
| | - Ki Su Kim
- School of Chemical Engineering, Pusan National University, Busan 46241, Republic of Korea; Department of Organic Materials Science and Engineering, Pusan National University, Busan 46241, Republic of Korea; Institute of Advanced Organic Materials, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
4
|
Pham DT, Thuy NTN, Thao NTP, Nhi LT, Thuy BTP. Naturally derived hydrogels for wound healing. Ther Deliv 2025; 16:349-363. [PMID: 39871586 PMCID: PMC11970767 DOI: 10.1080/20415990.2025.2457928] [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: 10/06/2024] [Accepted: 01/21/2025] [Indexed: 01/29/2025] Open
Abstract
Natural hydrogels have garnered increasing attention due to their natural origins and beneficial roles in wound healing. Hydrogel water-retaining capacity and excellent biocompatibility create an ideal moist environment for wound healing, thereby enhancing cell proliferation and tissue regeneration. For this reason, naturally derived hydrogels formulated from biomaterials such as chitosan, alginate, gelatin, and fibroin are highly promising due to their biodegradability and low immunogenic responses. Recent integrated approaches to utilizing new technologies with bioactive agents have significantly improved the mechanical properties of hydrogels and the controlled release and delivery of active compounds, thereby increasing the efficiency of the treatment processes. Herein, this review highlights the advantages and the challenges of natural hydrogels in wound healing, focusing on their mechanical strength, controlled degradation rates, safety and efficiency validation, and the potential for incorporating advanced technologies such as tissue engineering and gene therapy for utilization in personalized medicine.
Collapse
Affiliation(s)
- Duy Toan Pham
- Department of Health Sciences, College of Natural Sciences, Can Tho University, Can Tho, Vietnam
| | - Ngo Thi Ngoc Thuy
- Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Nguyen Thi Phuong Thao
- Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Le Thi Nhi
- Faculty of Materials Science, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Bui Thi Phuong Thuy
- Faculty of Fundamental Sciences, Van Lang University, Ho Chi Minh City, Vietnam
| |
Collapse
|
5
|
Yong R, Mu R, Han C, Chao T, Liu Y, Dong L, Wang C. Optimizing a 5-factor cocktail to prepare reparative macrophages for wound healing. J Leukoc Biol 2025; 117:qiae096. [PMID: 38630870 DOI: 10.1093/jleuko/qiae096] [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: 01/05/2024] [Revised: 03/14/2024] [Accepted: 04/06/2024] [Indexed: 04/19/2024] Open
Abstract
The treatment of nonhealing wounds, such as diabetic ulcers, remains a critical clinical challenge. Recent breakthroughs in cell therapy have shown great promise, with one primary focus on preparing cells with comprehensive reparative functions and foreseeable safety. In our previous study, we recapitulated the proregenerative and immunosuppressive functions of tumor-associated macrophages in non-tumor-derived macrophages, endowing the latter with characteristics for promoting diabetic wound healing-termed tumor-associated macrophage-educated macrophages. To eliminate the use of tumor-derived sources and devise a more controllable method to prepare tumor-associated macrophage-educated macrophage-like cells, in this study, we identify a cocktail comprising 5 recombinant proteins as an essential condition to induce nonpolarized macrophages into therapeutic cells with prohealing functions. The screened 5 factors are osteopontin, macrophage inflammatory protein 2, chemokine (C-C motif) ligand 8, vascular endothelial growth factor B, and macrophage colony-stimulating factor. We demonstrate the rationale for screening these factors and the phenotype of the 5 factor-induced tumor-associated macrophage-educated macrophage-like macrophages prepared from murine bone marrow-derived macrophages, which exhibit angiogenic and immunomodulatory effects in vitro. Then, we induce primary human monocytes from periphery blood into the 5 factor-induced tumor-associated macrophage-educated macrophage-like macrophages, which show prohealing effects in a human primary cell-based ex vivo model (T-Skin™). Our study demonstrates a simple, effective, and controllable approach to induce primary macrophages to possess repairing activities, which may provide insights for developing cell-based therapeutics for nonhealing wounds clinically.
Collapse
Affiliation(s)
- Rong Yong
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| | - Ruoyu Mu
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| | - Congwei Han
- School of Life Sciences & State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, No. 163 Xianlin Avenue, 210023, Nanjing, China
| | - Tzuwei Chao
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| | - Yu Liu
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| | - Lei Dong
- School of Life Sciences & State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, No. 163 Xianlin Avenue, 210023, Nanjing, China
- Chemistry and Biomedicine Innovative Center, Nanjing University, No. 163 Xianlin Avenue, 210023, Nanjing, Jiangsu, China
| | - Chunming Wang
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR, China
| |
Collapse
|
6
|
Yilmaz Ozdogan C, Kenar H, Uzuner H, Karadenizli A. Atelocollagen-based hydrogel loaded with Cotinus coggygriaextract for treatment of type 2 diabetic wounds. Biomed Mater 2025; 20:025009. [PMID: 39778335 DOI: 10.1088/1748-605x/ada7b5] [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: 06/13/2024] [Accepted: 01/08/2025] [Indexed: 01/11/2025]
Abstract
Diabetes, a chronic metabolic disease, causes complications such as chronic wounds, which are difficult to cure. New treatments have been investigated to accelerate wound healing. In this study, a novel wound dressing from fibroblast-laden atelocollagen-based hydrogel withCotinus coggygriaextract was developed for diabetic wound healing. The antimicrobial activity ofC. coggygriahexane (H), dichloromethane (DCM), dichloromethane:methanol (DCM-M), methanol (M), distilled water (DW) and traditional (T) extracts againstStaphylococcus aureus, Escherichia coli, Pseudomonas aeruginosa, Enterococcus faecalisandCandida albicans, as well as their cytotoxic effects on fibroblasts were determined. While fibroblast growth was significantly (p< 0.05) promoted with DCM (121.41 ± 1.04%), M (109.40 ± 5.89%) and DW (121.83 ± 6.37%) extracts at their lowest concentrations, 2000 μg ml-1DCM and 7.8 μg ml-1T extracts had both non-cytotoxic and antifungal effects. An atelocollagen-based hydrogel was produced by thermal crosslinking, and its pore size (38.75 ± 7.67 μm), water content (96.63 ± 0.24%) and swelling ratio (27.21 ± 4.08%) were found to be suitable for wound dressings. A significant increase in the deoxyribonucleic acid amount (28.27 ± 1.41%) was observed in the plain hydrogel loaded with fibroblasts after 9 d of incubation, and the hydrogel had an extensively interconnected cellular network. The hydrogels containing DW and T extracts were applied to wounds generated in anin vitro3D type-2-diabetic human skin model. Although the incubation period was not sufficient for closure of the wounds in either of the treatments, the hydrogel with T extract stimulated more fibroblast migration. In the fibroblast-laden version of the hydrogel with T extract, no wound closure was observed but more keratinocytes migrated to the wound region. These positive outcomes underline the potential of the developed wound dressing as a powerful alternative to improve diabetic wound healing in clinical practice.
Collapse
Affiliation(s)
- Candan Yilmaz Ozdogan
- Department of Medical Biology, Diabetes and Obesity Research Laboratory, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
- Department of Medical Microbiology, Molecular Research and Antibody Laboratory, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Halime Kenar
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
- ACU Biomaterials A & R Center, Acıbadem Mehmet Ali Aydınlar University (ACU), Istanbul, 34752, Turkey
| | - Huseyin Uzuner
- Department of Medical Microbiology, Molecular Research and Antibody Laboratory, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
- Department of Medical Services and Techniques, Kocaeli Vocational School of Health Services, Kocaeli University, Kocaeli, Turkey
| | - Aynur Karadenizli
- Department of Medical Microbiology, Molecular Research and Antibody Laboratory, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
7
|
Mansur AAP, Carvalho SM, Brito RMDM, Capanema NSV, Duval IDB, Cardozo ME, Rihs JBR, Lemos GGM, Lima LCD, dos Reys MP, Rodrigues APH, Oliveira LCA, de Sá MA, Cassali GD, Bueno LL, Fujiwara RT, Lobato ZIP, Mansur HS. Arginine-Biofunctionalized Ternary Hydrogel Scaffolds of Carboxymethyl Cellulose-Chitosan-Polyvinyl Alcohol to Deliver Cell Therapy for Wound Healing. Gels 2024; 10:679. [PMID: 39590035 PMCID: PMC11594054 DOI: 10.3390/gels10110679] [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: 09/19/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024] Open
Abstract
Wound healing is important for skin after deep injuries or burns, which can lead to hospitalization, long-term morbidity, and mortality. In this field, tissue-engineered skin substitutes have therapy potential to assist in the treatment of acute and chronic skin wounds, where many requirements are still unmet. Hence, in this study, a novel type of biocompatible ternary polymer hybrid hydrogel scaffold was designed and produced through an entirely eco-friendly aqueous process composed of carboxymethyl cellulose, chitosan, and polyvinyl alcohol and chemically cross-linked by citric acid, forming three-dimensional (3D) matrices, which were biofunctionalized with L-arginine (L-Arg) to enhance cellular adhesion. They were applied as bilayer skin biomimetic substitutes based on human-derived cell cultures of fibroblasts and keratinocytes were seeded and grown into their 3D porous structures, producing cell-based bio-responsive hybrid hydrogel scaffolds to assist the wound healing process. The results demonstrated that hydrophilic hybrid cross-linked networks were formed via esterification reactions with the 3D porous microarchitecture promoted by foam templating and freeze-drying. These hybrids presented chemical stability, physicochemical properties, high moisture adsorption capacity, surface properties, and a highly interconnected 3D porous structure well suited for use as a skin substitute in wound healing. Additionally, the surface biofunctionalization of these 3D hydrogel scaffolds with L-arginine through amide bonds had significantly enhanced cellular attachment and proliferation of fibroblast and keratinocyte cultures. Hence, the in vivo results using Hairless mouse models (an immunocompromised strain) confirmed that these responsive bio-hybrid hydrogel scaffolds possess hemocompatibility, bioadhesion, biocompatibility, adhesiveness, biodegradability, and non-inflammatory behavior and are capable of assisting the skin wound healing process.
Collapse
Affiliation(s)
- Alexandra A. P. Mansur
- Center of Nanoscience, Nanotechnology, and Innovation—CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais, UFMG, Av. Presidente Antônio Carlos, 6627—Escola de Engenharia, Bloco 2—Sala 2233, Belo Horizonte 31270-901, MG, Brazil; (A.A.P.M.); (S.M.C.); (N.S.V.C.); (G.G.M.L.)
| | - Sandhra M. Carvalho
- Center of Nanoscience, Nanotechnology, and Innovation—CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais, UFMG, Av. Presidente Antônio Carlos, 6627—Escola de Engenharia, Bloco 2—Sala 2233, Belo Horizonte 31270-901, MG, Brazil; (A.A.P.M.); (S.M.C.); (N.S.V.C.); (G.G.M.L.)
| | - Ramayana M. de M. Brito
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, UFMG, Belo Horizonte 31270-901, MG, Brazil; (R.M.d.M.B.); (I.d.B.D.); (M.E.C.); (J.B.R.R.); (L.L.B.); (R.T.F.)
| | - Nádia S. V. Capanema
- Center of Nanoscience, Nanotechnology, and Innovation—CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais, UFMG, Av. Presidente Antônio Carlos, 6627—Escola de Engenharia, Bloco 2—Sala 2233, Belo Horizonte 31270-901, MG, Brazil; (A.A.P.M.); (S.M.C.); (N.S.V.C.); (G.G.M.L.)
| | - Isabela de B. Duval
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, UFMG, Belo Horizonte 31270-901, MG, Brazil; (R.M.d.M.B.); (I.d.B.D.); (M.E.C.); (J.B.R.R.); (L.L.B.); (R.T.F.)
| | - Marcelo E. Cardozo
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, UFMG, Belo Horizonte 31270-901, MG, Brazil; (R.M.d.M.B.); (I.d.B.D.); (M.E.C.); (J.B.R.R.); (L.L.B.); (R.T.F.)
| | - José B. R. Rihs
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, UFMG, Belo Horizonte 31270-901, MG, Brazil; (R.M.d.M.B.); (I.d.B.D.); (M.E.C.); (J.B.R.R.); (L.L.B.); (R.T.F.)
| | - Gabriela G. M. Lemos
- Center of Nanoscience, Nanotechnology, and Innovation—CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais, UFMG, Av. Presidente Antônio Carlos, 6627—Escola de Engenharia, Bloco 2—Sala 2233, Belo Horizonte 31270-901, MG, Brazil; (A.A.P.M.); (S.M.C.); (N.S.V.C.); (G.G.M.L.)
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, UFMG, Belo Horizonte 31270-901, MG, Brazil; (R.M.d.M.B.); (I.d.B.D.); (M.E.C.); (J.B.R.R.); (L.L.B.); (R.T.F.)
| | - Letícia C. D. Lima
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, UFMG, Belo Horizonte 31270-901, MG, Brazil; (L.C.D.L.); (M.A.d.S.)
| | - Marina P. dos Reys
- Laboratory of Compared Pathology, Department of Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, UFMG, Belo Horizonte 31270-901, MG, Brazil; (M.P.d.R.); (G.D.C.)
| | - Ana P. H. Rodrigues
- Chemistry Department, Federal University of Minas Gerais, UFMG, Belo Horizonte 31270-901, MG, Brazil; (A.P.H.R.); (L.C.A.O.)
| | - Luiz C. A. Oliveira
- Chemistry Department, Federal University of Minas Gerais, UFMG, Belo Horizonte 31270-901, MG, Brazil; (A.P.H.R.); (L.C.A.O.)
| | - Marcos Augusto de Sá
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, UFMG, Belo Horizonte 31270-901, MG, Brazil; (L.C.D.L.); (M.A.d.S.)
| | - Geovanni D. Cassali
- Laboratory of Compared Pathology, Department of Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, UFMG, Belo Horizonte 31270-901, MG, Brazil; (M.P.d.R.); (G.D.C.)
| | - Lilian L. Bueno
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, UFMG, Belo Horizonte 31270-901, MG, Brazil; (R.M.d.M.B.); (I.d.B.D.); (M.E.C.); (J.B.R.R.); (L.L.B.); (R.T.F.)
| | - Ricardo T. Fujiwara
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, UFMG, Belo Horizonte 31270-901, MG, Brazil; (R.M.d.M.B.); (I.d.B.D.); (M.E.C.); (J.B.R.R.); (L.L.B.); (R.T.F.)
| | - Zelia I. P. Lobato
- Departamento de Medicina Veterinária Preventiva, Federal University of Minas Gerais, UFMG, Belo Horizonte 31270-901, MG, Brazil;
| | - Herman S. Mansur
- Center of Nanoscience, Nanotechnology, and Innovation—CeNano2I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais, UFMG, Av. Presidente Antônio Carlos, 6627—Escola de Engenharia, Bloco 2—Sala 2233, Belo Horizonte 31270-901, MG, Brazil; (A.A.P.M.); (S.M.C.); (N.S.V.C.); (G.G.M.L.)
| |
Collapse
|
8
|
Mozaffari N, Mohammadi R, Delirezh N, Hobbenaghi R, Mohammadi V. Effect of macrophages combined with supernatant of mesenchymal stem cell culture and macrophage culture on wound healing in rats. Tissue Cell 2024; 90:102474. [PMID: 39079451 DOI: 10.1016/j.tice.2024.102474] [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: 03/13/2024] [Revised: 06/22/2024] [Accepted: 07/11/2024] [Indexed: 09/03/2024]
Abstract
Wound healing is an orderly sequence of events restoring the integrity of the damaged tissue. It consists of inflammatory, proliferation, and remodeling phases. The objective of the current study was to investigate the effect of local transplantation of cultured macrophage loaded with mesenchymal stem cell/macrophage culture supernatants on wound healing. Sixty-four healthy adult male Wistar rats were randomized into 4 groups of sixteen animals each: 1) SHAM group. 2) MAC-MSC/SN group: One-milliliter application of a mixture comprising mesenchymal stem cell and macrophage culture supernatants in a 1:1 ratio was administered locally to the wound bed. 3) MAC group: Local transplantation of macrophage cells cultured in the wound bed. 4) MAC + MAC-MSC/SN group: Local transplantation of cultured macrophage in combination with mesenchymal stem cell/ macrophage culture supernatants in the wound bed. An incisional wound model was used for biomechanical studies, while an excisional wound model was used for biochemical, histopathological, and planimetric assessments. The wound area was significantly reduced in the MAC + MAC-MSC/SN group compared to other groups (P < 0.05). Biomechanical measurements from the MAC + MAC-MSC/SN group were significantly higher compared to other experimental groups (P < 0.05). Biochemical and quantitative histopathological analyses revealed a significant difference between MAC + MAC-MSC/SN and other groups (P < 0.05). MAC + MAC-MSC/SN showed the potential to improve wound healing significantly. This appears to work by angiogenesis stimulation, fibroblast proliferation, inflammation reduction, and granulation tissue formation during the initial stages of the healing process. This accelerated healing leads to earlier wound area reduction and enhanced tensile strength of the damaged area due to the reorganization of granulation tissue and collagen fibers.
Collapse
Affiliation(s)
- Nima Mozaffari
- Department of Surgery and Diagnostic Imaging, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Rahim Mohammadi
- Department of Surgery and Diagnostic Imaging, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.
| | - Nowruz Delirezh
- Department of Microbiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Rahim Hobbenaghi
- Department of Pathobiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Vahid Mohammadi
- Department of Internal Medicine and Clinical Pathology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| |
Collapse
|
9
|
Zhang X, Liang Y, Huang S, Guo B. Chitosan-based self-healing hydrogel dressing for wound healing. Adv Colloid Interface Sci 2024; 332:103267. [PMID: 39121832 DOI: 10.1016/j.cis.2024.103267] [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: 03/26/2024] [Revised: 06/02/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024]
Abstract
Skin has strong self-regenerative capacity, while severe skin defects do not heal without appropriate treatment. Therefore, in order to cover the wound sites and hasten the healing process, wound dressings are required. Hydrogels have emerged as one of the most promising candidates for wound dressings because of their hydrated and porous molecular structure. Chitosan (CS) with biocompatibility, oxygen permeability, hemostatic and antimicrobial properties is beneficial for wound treatment and it can generate self-healing hydrogels through reversible crosslinks, from dynamic covalent bonding, such as Schiff base bonds, boronate esters, and acylhydrazone bonds, to physical interactions like hydrogen bonding, electrostatic interaction, ionic bonding, metal-coordination, host-guest interactions, and hydrophobic interaction. Therefore, various chitosan-based self-healing hydrogel dressings have been prepared in recent years to cope with increasingly complex wound conditions. This review's objective is to provide comprehensive information on the self-healing mechanism of chitosan-based hydrogel wound dressings, discuss their advanced functions including antibacterial, conductive, anti-inflammatory, anti-oxidant, stimulus-responsive, hemostatic/adhesive and controlled release properties, further introduce their applications in the promotion of wound healing in two categories: acute and chronic (infected, burn and diabetic) wounds, and finally discuss the future perspective of chitosan-based self-healing hydrogel dressings for wound healing.
Collapse
Affiliation(s)
- Xingyu Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710049, China; State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yongping Liang
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Shengfei Huang
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Baolin Guo
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710049, China; State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
10
|
Shimizu Y, Ntege EH, Inoue Y, Matsuura N, Sunami H, Sowa Y. Optimizing mesenchymal stem cell extracellular vesicles for chronic wound healing: Bioengineering, standardization, and safety. Regen Ther 2024; 26:260-274. [PMID: 38978963 PMCID: PMC11228664 DOI: 10.1016/j.reth.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/12/2024] [Accepted: 06/06/2024] [Indexed: 07/10/2024] Open
Abstract
Chronic wounds represent a significant global burden, afflicting millions with debilitating complications. Despite standard care, impaired healing persists due to factors like persistent inflammation and impaired tissue regeneration. Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) offer an innovative regenerative medicine approach, delivering stem cell-derived therapeutic cargo in engineered nanoscale delivery systems. This review examines pioneering bioengineering strategies to engineer MSC-EVs into precision nanotherapeutics for chronic wounds. Emerging technologies like CRISPR gene editing, microfluidic manufacturing, and biomimetic delivery systems are highlighted for their potential to enhance MSC-EV targeting, optimize therapeutic cargo enrichment, and ensure consistent clinical-grade production. However, key hurdles remain, including batch variability, rigorous safety assessment for potential tumorigenicity, immunogenicity, and biodistribution profiling. Crucially, collaborative frameworks harmonizing regulatory science with bioengineering and patient advocacy hold the key to expediting global clinical translation. By overcoming these challenges, engineered MSC-EVs could catalyze a new era of off-the-shelf regenerative therapies, restoring hope and healing for millions afflicted by non-healing wounds.
Collapse
Affiliation(s)
- Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Edward Hosea Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Yoshikazu Inoue
- Department of Plastic and Reconstructive Surgery, School of Medicine, Fujita Health University, 1-98, Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Naoki Matsuura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Hiroshi Sunami
- Center for Advanced Medical Research, School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, 329-0498, Tochigi, Japan
| |
Collapse
|
11
|
Ertlen C, Seblani M, Bonnet M, Brezun JM, Coyle T, Sabatier F, Fuentes S, Decherchi P, Serratrice N, Marqueste T. Efficacy of the immediate adipose-derived stromal vascular fraction autograft on functional sensorimotor recovery after spinal cord contusion in rats. Stem Cell Res Ther 2024; 15:29. [PMID: 38303017 PMCID: PMC10835949 DOI: 10.1186/s13287-024-03645-z] [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: 09/13/2023] [Accepted: 01/23/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Spinal cord injuries (SCI) lead to functional alteration with important consequences such as motor and sensory disorders. The repair strategies developed to date remain ineffective. The adipose tissue-derived stromal vascular fraction (SVF) is composed of a cocktail of cells with trophic, pro-angiogenic and immunomodulatory effects. Numerous therapeutic benefits were shown for tissue reconstitution, peripheral neuropathy and for the improvement of neurodegenerative diseases. Here, the therapeutic efficacy of SVF on sensorimotor recovery after an acute thoracic spinal cord contusion in adult rats was determined. METHOD Male Sprague Dawley rats (n = 45) were divided into 3 groups: SHAM (without SCI and treatment), NaCl (animals with a spinal lesion and receiving a saline injection through the dura mater) and SVF (animals with a spinal lesion and receiving a fraction of fat removed from adipocytes through the dura mater). Some animals were sacrificed 14 days after the start of the experiment to determine the inflammatory reaction by measuring the interleukin-1β, interleukin-6 and Tumor Necrosis Factor-α in the lesion area. Other animals were followed once a week for 12 weeks to assess functional recovery (postural and locomotor activities, sensorimotor coordination). At the end of this period, spinal reflexivity (rate-dependent depression of the H-reflex) and physiological adjustments (ventilatory response to metabosensitive muscle activation following muscle fatigue) were measured with electrophysiological tools. RESULTS Compared to non-treated animals, results indicated that the SVF reduced the endogenous inflammation and increased the behavioral recovery in treated animals. Moreover, H-reflex depression and ventilatory adjustments to muscle fatigue were found to be comparable between SHAM and SVF groups. CONCLUSION Our results highlight the effectiveness of SVF and its high therapeutic potential to improve sensorimotor functions and to restore the segmental sensorimotor loop and the communication between supra- and sub-lesional spinal cord regions after traumatic contusion.
Collapse
Affiliation(s)
- Céline Ertlen
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - Mostafa Seblani
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - Maxime Bonnet
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - Jean-Michel Brezun
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - Thelma Coyle
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - Florence Sabatier
- Assistance Publique - Hôpitaux de Marseille (AP-HM), INSERM 1409 Centre d'Investigation Clinique en Biothérapies, Unité de Culture Et Thérapie Cellulaire, Hôpital de La Conception, 147, Boulevard Baille, 13385, Marseille Cedex 05, France
| | - Stéphane Fuentes
- Assistance Publique - Hôpitaux de Marseille (AP-HM), Service de Neurochirurgie, Hôpital de La Timone, 264, Rue Saint-Pierre, 13005, Marseille, France
| | - Patrick Decherchi
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France.
| | - Nicolas Serratrice
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
- Assistance Publique - Hôpitaux de Marseille (AP-HM), Service de Neurochirurgie, Hôpital de La Timone, 264, Rue Saint-Pierre, 13005, Marseille, France
| | - Tanguy Marqueste
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France.
| |
Collapse
|
12
|
Shahmohammadi A, Samadian H, Heidari Keshel S, Rashidi K, Kiani A, Soleimani M, Goudarzi F. Burn wound healing using adipose-derived mesenchymal stem cells and manganese nanoparticles in polycaprolactone/gelatin electrospun nanofibers in rats. BIOIMPACTS : BI 2024; 14:30193. [PMID: 39296800 PMCID: PMC11406429 DOI: 10.34172/bi.2024.30193] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/01/2024] [Accepted: 01/02/2024] [Indexed: 09/21/2024]
Abstract
Introduction Wound healing is a major therapeutic concern in regenerative medicine. The current study aimed to investigate the second-degree burn wound treatment in rats using rat adipose- derived stem cells (ADSCs) and manganese nanoparticles (MnO2-NPs) in a polycaprolactone/gelatin electrospun nanofiber scaffold. Methods After the synthesis of nanoparticles and electrospinning of nanofibers, the SEM analysis, contact angle, mechanical strength, blood compatibility, porosity, swelling, biodegradability, cell viability, and adhesion assays were performed. According to the results, the PCL/Gel/5%MnO2-NPs nanofiber (Mn-5%) was determined to be the most suitable scaffold. The ADSCs-seeded Mn-5% scaffolds were applied as a burn wound dressing. The wound closure rate, IL-1β, and IL-6 level, hydroxyproline, and glycosaminoglycans content were measured, and the hematoxylin and eosin, Masson's trichrome, and immunohistochemistry stainings were carried out. Results Based on the results, in Mn+S (ADSCs+PCL/Gel/5%MnO2-NPs nanofiber) and N+S (ADSCs+PCL/Gel nanofiber) groups, the IL-6 and IL-1β levels were reduced, and the percentage of wound closure, glycosaminoglycans, and hydroxyproline content were increased compared to the control group (P<0.05). Also, the lowest amount of α-SMA was observed in these two groups, demonstrating stem cells' role in reducing α-SMA levels and thus preventing fibrosis. Moreover, the amount of α-SMA in the Mn+S group is lower than in the N+S group and, is closer to healthy skin. According to histology results, the best type of treatment was observed in the Mn+S group. Conclusion In conclusion, the ADSCs-seeded PCL/Gel/5%MnO2-NPs scaffold demonstrated considerable therapeutic effects in burn wound healing.
Collapse
Affiliation(s)
- Azin Shahmohammadi
- Department of Applied Cell Sciences and Tissue Engineering, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadi Samadian
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saeed Heidari Keshel
- Department of Applied Cell Sciences and Tissue Engineering, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khodabakhsh Rashidi
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amir Kiani
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Soleimani
- Department of Applied Cell Sciences and Tissue Engineering, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farjam Goudarzi
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
13
|
Dalal N, Challa R, Thimukonda JJ, Tayalia P. Gelatin Methacryloyl Based Injectable Cryogels with Tunable Degradability for Cell Delivery. Macromol Biosci 2024; 24:e2200562. [PMID: 36974501 DOI: 10.1002/mabi.202200562] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/09/2023] [Indexed: 03/29/2023]
Abstract
Scaffold-based cell delivery can improve therapeutic effects of transplanted cells in cell therapy. Biomaterial scaffolds serveas niche for cell growth and proliferation which improves cell survival and overall function post cell delivery. In this study, gelatin methacryloyl based injectable scaffolds made using poly(ethylene)glycol as a sacrificial polymer and cryogelation as a technique, are demonstrated to have tunable degradability and porosity that is required for cell and drug delivery applications. The pore size (10-142 µm) of these gels makes them suitable for loading different cell types as per the application. In vitro studies using mammalian cells confirm that these cryogels are cytocompatible. These cell-laden scaffolds are injectable and have a cell retention ability of up to 90% after injection. Rheology is done to evaluate stiffness and shape recovery property, and it is found that these gels can maintain their original shape even after applying 7 cycles of strain from 0.1% to 20%. Furthermore, their degradability can be modulated between 6 and 10 days by changing the overall polymer composition. Thus, injectability and degradability of these cryogels can circumvent invasive surgical procedures, thereby making them useful for a variety of applications including delivery of cells and bioactive factors.
Collapse
Affiliation(s)
- Neha Dalal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Ramadevi Challa
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Jeyapriya J Thimukonda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Prakriti Tayalia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| |
Collapse
|
14
|
Biscari G, Fan Y, Namata F, Fiorica C, Malkoch M, Palumbo FS, Pitarresi G. Antibacterial Broad-Spectrum Dendritic/Gellan Gum Hybrid Hydrogels with Rapid Shape-Forming and Self-Healing for Wound Healing Application. Macromol Biosci 2023; 23:e2300224. [PMID: 37590124 DOI: 10.1002/mabi.202300224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/31/2023] [Indexed: 08/19/2023]
Abstract
Treating wound infections is a difficult task ever since pathogenic bacteria started to develop resistance to common antibiotics. The present study develops hybrid hydrogels based on the formation of a polyelectrolyte complex between the anionic charges of dopamine-functionalized Gellan Gum (GG-DA) and the cationic moieties of the TMP-G2-alanine dendrimer. The hydrogels thus obtained can be doubly crosslinked with CaCl2 , obtaining solid hydrogels. Or, by oxidizing dopamine to GG-DA, possibly causing further interactions such as Schiff Base and Michael addition to take place, hydrogels called injectables can be obtained. The latter have shear-thinning and self-healing properties (efficiency up to 100%). Human dermal fibroblasts (HDF), human epidermal keratinocytes (HaCaT), and mouse monocyte cells (RAW 264.7), after incubation with hydrogels, in most cases show cell viability up to 100%. Hydrogels exhibit adhesive behavior on various substrates, including porcine skin. At the same time, the dendrimer serves to crosslink the hydrogels and endows them with excellent broad-spectrum microbial eradication activity within four hours, evaluated using Staphylococcus aureus 2569 and Escherichia coli 178. Using the same GG-DA/TMP-G2-alanine ratios hybrid hydrogels with tunable properties and potential for wound dressing applications can be produced.
Collapse
Affiliation(s)
- Giuseppina Biscari
- KTH Royal Institute of Technology, Teknikringen 56-58, Stockholm, SE-100 44, Sweden
| | - Yanmiao Fan
- University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Faridah Namata
- University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | - Calogero Fiorica
- KTH Royal Institute of Technology, Teknikringen 56-58, Stockholm, SE-100 44, Sweden
| | - Michael Malkoch
- University of Palermo, Via Archirafi 32, Palermo, 90123, Italy
| | | | - Giovanna Pitarresi
- KTH Royal Institute of Technology, Teknikringen 56-58, Stockholm, SE-100 44, Sweden
| |
Collapse
|
15
|
Mbituyimana B, Adhikari M, Qi F, Shi Z, Fu L, Yang G. Microneedle-based cell delivery and cell sampling for biomedical applications. J Control Release 2023; 362:692-714. [PMID: 37689252 DOI: 10.1016/j.jconrel.2023.09.013] [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: 05/19/2023] [Revised: 08/16/2023] [Accepted: 09/04/2023] [Indexed: 09/11/2023]
Abstract
Cell-based therapeutics are novel therapeutic strategies that can potentially treat many presently incurable diseases through novel mechanisms of action. Cell therapies may benefit from the ease, safety, and efficacy of administering therapeutic cells. Despite considerable recent technological and biological advances, several barriers remain to the clinical translation and commercialization of cell-based therapies, including low patient compliance, personal handling inconvenience, poor biosafety, and limited biocompatibility. Microneedles (MNs) are emerging as a promising biomedical device option for improved cell delivery with little invasion, pain-free administration, and simplicity of disposal. MNs have shown considerable promise in treating a wide range of diseases and present the potential to improve cell-based therapies. In this review, we first summarized the latest advances in the various types of MNs developed for cell delivery and cell sampling. Emphasis was given to the design and fabrication of various types of MNs based on their structures and materials. Then we focus on the recent biomedical applications status of MNs-mediated cell delivery and sampling, including tissue repair (wound healing, heart repair, and endothelial repair), cancer treatment, diabetes therapy, cell sampling, and other applications. Finally, the current status of clinical application, potential perspectives, and the challenges for clinical translation are also highlighted.
Collapse
Affiliation(s)
- Bricard Mbituyimana
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Manjila Adhikari
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Fuyu Qi
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhijun Shi
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Lina Fu
- College of Medicine, Huanghuai University, Zhumadian, Henan 463000, China; Zhumadian Central Hospital, Zhumadian, Henan 463000, China.
| | - Guang Yang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
16
|
Riccio M, Bondioli E, Senesi L, Zingaretti N, Gargiulo P, De Francesco F, Parodi PC, Zavan B. Fragmented Dermo-Epidermal Units (FdeU) as an Emerging Strategy to Improve Wound Healing Process: An In Vitro Evaluation and a Pilot Clinical Study. J Clin Med 2023; 12:6165. [PMID: 37834809 PMCID: PMC10573238 DOI: 10.3390/jcm12196165] [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: 07/07/2023] [Revised: 09/06/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Innovative strategies have shown beneficial effects in healing wound management involving, however, a time-consuming and arduous process in clinical contexts. Micro-fragmented skin tissue acts as a slow-released natural scaffold and continuously delivers growth factors, and much other modulatory information, into the microenvironment surrounding damaged wounds by a paracrine function on the resident cells which supports the regenerative process. In this study, in vitro and in vivo investigations were conducted to ascertain improved effectiveness and velocity of the wound healing process with the application of fragmented dermo-epidermal units (FdeU), acquired via a novel medical device (Hy-Tissue® Micrograft Technology). MTT test; LDH test; ELISA for growth factor investigation (IL) IL-2, IL-6, IL-7 IL-8, IL-10; IGF-1; adiponectin; Fibroblast Growth Factor (FGF); Vascular Endothelial Growth Factor (VEGF); and Tumor Necrosis Factor (TNF) were assessed. Therefore, clinical evaluation in 11 patients affected by Chronic Wounds (CW) and treated with FdeU were investigated. Functional outcome was assessed pre-operatory, 2 months after treatment (T0), and 6 months after treatment (T1) using the Wound Bed Score (WBS) and Vancouver Scar Scale (VSS). In this current study, we demonstrate the potential of resident cells to proliferate from the clusters of FdeU seeded in a monolayer that efficiently propagate the chronic wound. Furthermore, in this study we report how the discharge of trophic/reparative proteins are able to mediate the in vitro paracrine function of proliferation, migration, and contraction rate in fibroblasts and keratinocytes. Our investigations recommend FdeU as a favorable tool in wound healing, displaying in vitro growth-promoting potential to enhance current therapeutic mechanisms.
Collapse
Affiliation(s)
- Michele Riccio
- Department of Reconstructive Surgery and Hand Surgery, University Hospital (AOU Ospedali Riuniti di Ancona), Via Conca 71, Torrette di Ancona, 60123 Ancona, Italy; (M.R.); (L.S.); (F.D.F.)
| | - Elena Bondioli
- Burn Center and Emilia Romagna Regional Skin Bank, Bufalini Hospital, AUSL della Romagna, 47521 Cesena, Italy;
| | - Letizia Senesi
- Department of Reconstructive Surgery and Hand Surgery, University Hospital (AOU Ospedali Riuniti di Ancona), Via Conca 71, Torrette di Ancona, 60123 Ancona, Italy; (M.R.); (L.S.); (F.D.F.)
| | - Nicola Zingaretti
- Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy; (N.Z.); (P.C.P.)
| | - Paolo Gargiulo
- Engineering Department, King’s College, London WC2R 2LS, UK;
- Institute for Biomedical and Neural Engineering, Reykjavík University, 101 Reykjavík, Iceland
| | - Francesco De Francesco
- Department of Reconstructive Surgery and Hand Surgery, University Hospital (AOU Ospedali Riuniti di Ancona), Via Conca 71, Torrette di Ancona, 60123 Ancona, Italy; (M.R.); (L.S.); (F.D.F.)
| | - Pier Camillo Parodi
- Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy; (N.Z.); (P.C.P.)
| | - Barbara Zavan
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy
| |
Collapse
|
17
|
Namgoong S, Baik S, Han SK, Son JW, Kim JY. Developing and Establishing a Wound Dressing Team: Experience and Recommendations. J Korean Med Sci 2023; 38:e168. [PMID: 37270921 DOI: 10.3346/jkms.2023.38.e168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 02/16/2023] [Indexed: 06/06/2023] Open
Abstract
BACKGROUND The existing literature has comprehensively examined the benefits of specialized wound-care services and multidisciplinary team care. However, information on the development and integration of wound-dressing teams for patients who do not require specialized wound care is scarce. Therefore, the present study aimed to elucidate the benefits of a wound-dressing team by reporting our experiences with the establishment of a wound-dressing team. METHODS The wound-dressing team was established at Korea University Guro Hospital. Between July 2018 and June 2022, 180,872 cases were managed for wounds at the wound-dressing team. The data were analyzed to assess the types of wounds and their outcomes. In addition, questionnaires assessing the satisfaction with the service were administered to patients, ward nurses, residents/internists, and team members. RESULTS Regarding the wound type, 80,297 (45.3%) were catheter-related, while 48,036 (27.1%), 26,056 (14.7%), and 20,739 (11.7%) were pressure ulcers, dirty wounds, and simple wounds, respectively. In the satisfaction survey, the scores of the patient, ward nurse, dressing team nurse, and physician groups were 8.9, 8.1, 8.2, and 9.1, respectively. Additionally, 136 dressing-related complications (0.08%) were reported. CONCLUSION The wound dressing team can enhance satisfaction among patients and healthcare providers with low complications. Our findings may provide a potential framework for establishing similar service models.
Collapse
Affiliation(s)
- Sik Namgoong
- Department of Plastic Surgery, Korea University College of Medicine, Seoul, Korea
- Diabetic Wound Center, Korea University Guro Hospital, Seoul, Korea
| | - Seunghee Baik
- Department of Plastic Surgery, Korea University College of Medicine, Seoul, Korea
| | - Seung-Kyu Han
- Department of Plastic Surgery, Korea University College of Medicine, Seoul, Korea
- Diabetic Wound Center, Korea University Guro Hospital, Seoul, Korea.
| | - Ji-Won Son
- Diabetic Wound Center, Korea University Guro Hospital, Seoul, Korea
- Department of Nursing Service, Korea University Guro Hospital, Seoul, Korea
| | - Jae-Yeon Kim
- Department of Nursing Service, Korea University Guro Hospital, Seoul, Korea
| |
Collapse
|
18
|
Karakol P, Bozkurt M, Gelbal C, Tuglu MI. Efficacy of stromal vascular fraction and enzyme-free mechanical isolation therapy in experimental full thickness burn wounds. J Plast Surg Hand Surg 2023; 57:78-94. [PMID: 34709935 DOI: 10.1080/2000656x.2021.1993234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Autologous cell suspensions obtained by a stromal vascular fraction (SVF) and enzyme-free mechanical isolation (EMI) are an alternative in the treatment of burn wounds. In this study, we aimed to investigate the effect of autologous cell suspensions obtained by SVF and EMI on full-thickness skin burn wounds. METHODS A total of 45 male Sprague-Dawley rats were divided into three groups, SVF group, EMI group, and SVF + EMI group. The groups were also classified as the first, second, and third week of the burn to reveal the effect of the treatment on the burn in the early, middle, and late stages. For treatment, 0.2 ml SVF or 0.2 ml EMI was injected subcutaneously into the burn lesions of the subjects. Histopathological examination was performed on the burn wounds taken at the end of the experiment, and Ki67, CD44, CD73, CD90, and CK17 expressions were evaluated. RESULTS Histological examination revealed that there was no improvement in the control samples, but the skin was multicellular, vascularization was present. Histologic scores in all groups was significantly better than control, and SVF + EMI was the best group in terms of recovery (p < 0.05). Ki67, CK17, CD44, CD73, and CD90 expressions were significantly higher in the treatment groups compared to the control (p < 0.05). CONCLUSION We found in our study that both applications significantly increased the healing of the burn wound. Moreover, SVF + EMI application provided more improvement than SVF or EMI alone.
Collapse
Affiliation(s)
- Percin Karakol
- Department of Plastic, Reconstructive and Aesthetic Surgery, Health Science University Bağcilar Education and Training Hospital, Istanbul, Turkey
| | - Mehmet Bozkurt
- Department of Plastic, Reconstructive and Aesthetic Surgery, Health Science University Bağcilar Education and Training Hospital, Istanbul, Turkey
| | - Caner Gelbal
- Department of Plastic, Reconstructive and Aesthetic Surgery, Health Science University Bağcilar Education and Training Hospital, Istanbul, Turkey
| | - Mehmet Ibrahim Tuglu
- Faculty of Medicine, Department of Histology, Celal Bayar University, Manisa, Turkey
| |
Collapse
|
19
|
Yang P, Zhang S, Yan T, Li F, Zhang S. The Therapeutic Application of Stem Cells and Their Derived Exosomes in the Treatment of Radiation-Induced Skin Injury. Radiat Res 2023; 199:182-201. [PMID: 36630584 DOI: 10.1667/rade-22-00023.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 12/05/2022] [Indexed: 01/13/2023]
Abstract
Radiation-induced skin injury (RISI) is a serious concern for nuclear accidents and cancer radiotherapy, which seriously affects the quality of life of patients. This injury differs from traditional wounds due to impaired healing and the propensity to recurrence and is divided into acute and chronic phases on the basis of the injury time. Unfortunately, there are few effective therapies for preventing or mitigating this injury. Over the last few decades, various studies have focused on the effects of stem cell-based therapies to address the tissue repair and regeneration of irradiated skin. These stem cells modulate inflammation and instigate tissue repair by differentiating into specific kinds of cells or releasing paracrine factors. Stem cell-based therapies, including bone marrow-derived stem cells (BMSCs), adipose-derived stem cells (ADSCs) and stromal vascular fraction (SVF), have been reported to facilitate wound healing after radiation exposure. Moreover, stem cell-derived exosomes have recently been suggested as an effective and cell-free approach to support skin regeneration, circumventing the concerns respecting direct application of stem cells. Based on the literature on stem cell-based therapies for radiation-induced skin injury, we summarize the characteristics of different stem cells and describe their latest animal and clinical applications, as well as potential mechanisms. The promise of stem-cell based therapies against radiation-induced skin injury contribute to our response to nuclear events and smooth progress of cancer radiotherapy.
Collapse
Affiliation(s)
- Ping Yang
- Laboratory of Radiation Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Shuaijun Zhang
- Laboratory of Radiation Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Yan
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China.,Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China
| | - Fengsheng Li
- PLA Rocket Rorce Characteristic Medical Center, Beijing 100088, China
| | - Shuyu Zhang
- Laboratory of Radiation Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China.,Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China.,NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang 621099, China
| |
Collapse
|
20
|
Zhao X, Li X, Wang Y, Guo Y, Huang Y, Lv D, Lei M, Yu S, Luo G, Zhan R. Stability and biosafety of human epidermal stem cell for wound repair: preclinical evaluation. Stem Cell Res Ther 2023; 14:4. [PMID: 36600269 PMCID: PMC9814209 DOI: 10.1186/s13287-022-03202-6] [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: 05/24/2022] [Accepted: 07/20/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Cell therapy is a key technology to prevent sacrificing normal skin. Although some studies have shown the promise of human epidermal stem cells (EpiSCs), the efficacy, biosafety and quality control of EpiSC therapy have not been systematically reported. METHODS The biosafety, stemness maintenance and wound repair of EpiSC were systematically verified by in vitro and in vivo experiments. EpiSC were prepared from the foreskin using a collagen type IV rapid adherence method. The EpiSCs were identified by flow cytometry, immunofluorescence staining and cell morphology. The well-growing passage 1 (P1) EpiSCs were used to determine the proliferation curve (counting method). EpiSC clone formation assay was performed by Giemsa staining. Nude mice were used to prepare a full-thickness skin defect wound model to detect the repair effect of EpiSCs. The biosafety of EpiSCs was double tested in vitro and in vivo. RESULTS The results showed that the expression of specific markers and clone formation efficiency was stable when passage 1 (P1) to P8 cells were cultured, and the stemness rate of P8 cells was close to 85.1%. EpiSCs were expanded in vitro for 25 days, the number of cells reached 2.5 × 108, and the transplantable area was approximately 75% of the total body surface area (TBSA). At 45 days, the total number of cells was approximately 30 billion, and the transplantable area was approximately the size of a volleyball court. A nude mouse wound model indicated that EpiSCs could rapidly close a wound. On postinjury day 7, the wound epithelialization rate in the cell transplantation group was significantly higher than that in the NaCl group (P < 0.05). In vitro, cell senescence increased, and telomerase activity decreased in P1 to P8 EpiSCs. In vivo, there were no solid tumors or metastatic tumors after EpiSC (P8) transplantation. In addition, the quality control of cultured cells met the clinical application criteria for cell therapy. CONCLUSION This preclinical study showed the stability and biosafety of human EpiSC therapy for wound repair.
Collapse
Affiliation(s)
- Xiaohong Zhao
- grid.410570.70000 0004 1760 6682Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Xue Li
- grid.410570.70000 0004 1760 6682Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Ying Wang
- grid.410570.70000 0004 1760 6682Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Yicheng Guo
- grid.410570.70000 0004 1760 6682Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Yong Huang
- grid.410570.70000 0004 1760 6682Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Dalun Lv
- grid.452929.10000 0004 8513 0241Department of Burn and Plastic Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001 Anhui China
| | - Mingxing Lei
- grid.190737.b0000 0001 0154 0904“111” Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044 China
| | - Shicang Yu
- grid.410570.70000 0004 1760 6682Stem Cell and Regenerative Medicine, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038 China
| | - Gaoxing Luo
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Rixing Zhan
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
21
|
Hosseini M, Dalley AJ, Shafiee A. Convergence of Biofabrication Technologies and Cell Therapies for Wound Healing. Pharmaceutics 2022; 14:pharmaceutics14122749. [PMID: 36559242 PMCID: PMC9785239 DOI: 10.3390/pharmaceutics14122749] [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: 10/28/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cell therapy holds great promise for cutaneous wound treatment but presents practical and clinical challenges, mainly related to the lack of a supportive and inductive microenvironment for cells after transplantation. Main: This review delineates the challenges and opportunities in cell therapies for acute and chronic wounds and highlights the contribution of biofabricated matrices to skin reconstruction. The complexity of the wound healing process necessitates the development of matrices with properties comparable to the extracellular matrix in the skin for their structure and composition. Over recent years, emerging biofabrication technologies have shown a capacity for creating complex matrices. In cell therapy, multifunctional material-based matrices have benefits in enhancing cell retention and survival, reducing healing time, and preventing infection and cell transplant rejection. Additionally, they can improve the efficacy of cell therapy, owing to their potential to modulate cell behaviors and regulate spatiotemporal patterns of wound healing. CONCLUSION The ongoing development of biofabrication technologies promises to deliver material-based matrices that are rich in supportive, phenotype patterning cell niches and are robust enough to provide physical protection for the cells during implantation.
Collapse
Affiliation(s)
- Motaharesadat Hosseini
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia
- ARC Industrial Transformation Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing (M3D), Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Andrew J. Dalley
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD 4029, Australia
- Royal Brisbane and Women’s Hospital, Metro North Hospital and Health Service, Brisbane, QLD 4029, Australia
| | - Abbas Shafiee
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD 4029, Australia
- Royal Brisbane and Women’s Hospital, Metro North Hospital and Health Service, Brisbane, QLD 4029, Australia
- Frazer Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
- Correspondence: or
| |
Collapse
|
22
|
Cialdai F, Risaliti C, Monici M. Role of fibroblasts in wound healing and tissue remodeling on Earth and in space. Front Bioeng Biotechnol 2022; 10:958381. [PMID: 36267456 PMCID: PMC9578548 DOI: 10.3389/fbioe.2022.958381] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022] Open
Abstract
Wound healing (WH) and the role fibroblasts play in the process, as well as healing impairment and fibroblast dysfunction, have been thoroughly reviewed by other authors. We treat these topics briefly, with the only aim of contextualizing the true focus of this review, namely, the microgravity-induced changes in fibroblast functions involved in WH. Microgravity is a condition typical of spaceflight. Studying its possible effects on fibroblasts and WH is useful not only for the safety of astronauts who will face future interplanetary space missions, but also to help improve the management of WH impairment on Earth. The interesting similarity between microgravity-induced alterations of fibroblast behavior and fibroblast dysfunction in WH impairment on Earth is highlighted. The possibility of using microgravity-exposed fibroblasts and WH in space as models of healing impairment on Earth is suggested. The gaps in knowledge on fibroblast functions in WH are analyzed. The contribution that studies on fibroblast behavior in weightlessness can make to fill these gaps and, consequently, improve therapeutic strategies is considered.
Collapse
|
23
|
Lana JFSD, Lana AVSD, da Fonseca LF, Coelho MA, Marques GG, Mosaner T, Ribeiro LL, Azzini GOM, Santos GS, Fonseca E, de Andrade MAP. Stromal Vascular Fraction for Knee Osteoarthritis - An Update. J Stem Cells Regen Med 2022; 18:11-20. [PMID: 36003656 DOI: 10.46582/jsrm.1801003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 12/10/2021] [Indexed: 11/19/2022]
Abstract
Orthobiologics never cease to cause popularity within the medical science field, distinctly in regenerative medicine. Recently, adipose tissue has been an object of interest for many researchers and medical experts due to the fact that it represents a novel and potential cell source for tissue engineering and regenerative medicine purposes. Stromal vascular fraction (SVF), for instance, which is an adipose tissue-derivative, has generated optimistic results in many scenarios. Its biological potential can be harnessed and administered into injured tissues, particularly areas in which standard healing is disrupted. This is a typical feature of osteoarthritis (OA), a common degenerative joint disease which is outlined by persistent inflammation and destruction of surrounding tissues. SVF is known to carry a large amount of stem and progenitor cells, which are able to perform self-renewal, differentiation, and proliferation. Furthermore, they also secrete several cytokines and several growth factors, effectively sustaining immune modulatory effects and halting the escalated pro-inflammatory status of OA. Although SVF has shown interesting results throughout the medical community, additional research is still highly desirable in order to further elucidate its potential regarding musculoskeletal disorders, especially OA.
Collapse
Affiliation(s)
| | | | - Lucas Furtado da Fonseca
- Orthopaedic Department - Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo - SP, Brazil
| | - Marcelo Amaral Coelho
- IOC - Instituto do Osso e da Cartilagem / The Bone and Cartilage Institute, Indaiatuba - SP, Brazil
| | | | - Tomas Mosaner
- IOC - Instituto do Osso e da Cartilagem / The Bone and Cartilage Institute, Indaiatuba - SP, Brazil
| | | | | | - Gabriel Silva Santos
- IOC - Instituto do Osso e da Cartilagem / The Bone and Cartilage Institute, Indaiatuba - SP, Brazil
| | - Eduardo Fonseca
- IOC - Instituto do Osso e da Cartilagem / The Bone and Cartilage Institute, Indaiatuba - SP, Brazil
| | | |
Collapse
|
24
|
Zaccaron RP, Barbieri RT, Mendes C, Venturini LM, Alves N, Mariano SDS, de Andrade TAM, Hermes de Araújo PH, Feuser PE, Thirupathi A, Machado-de-Ávila RA, Lock Silveira PC. Photobiomodulation associated with lipid nanoparticles and hyaluronic acid accelerate the healing of excisional wounds. J Biomater Appl 2022; 37:668-682. [PMID: 35705485 DOI: 10.1177/08853282221109344] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objectives: This article aimed to investigate the effects of the association between photobiomodulation and hyaluronic acid incorporated in lipid nanoparticles in an epithelial lesion model in inflammatory parameters and oxidative stress. Methods: Eighty Wistar rats were randomly assigned to the following groups: epithelial lesion group (EL); EL+PBM; EL+HA; EL+SLNs; EL+SLNs-HA; EL+PBM+HA; EL+PBM+SLNs; EL+PBM+SLNs-HA. The animals were anesthetized with 4% isofluorane after shaving and induced to an epithelial lesion. Topical treatment with a gel containing HA (0.9%) and/or SLNs (10 mg/mL) and with laser irradiation occurred daily for 1 week. Results: The results showed an increase in wound contraction on the seventh day in the LE + LBM + AH-NPL group, a reduction in pro-inflammatory cytokines (IL-6, IL-1β, and TNF-α), an increase in anti-inflammatory cytokines (IL- 4 and IL-10) and TGF-β. The levels of pro-inflammatory cytokine IL-4 and TGF-β also showed an increase in the LE + NPL-AH, LE + FBM + AH, LE + FBM + NPL and LE + FBM + NPL-AH groups. Regarding oxidative stress parameters, the levels of DCF and nitrite decreased in the combined therapy group when compared to the control group, as well as oxidative damage (carbonyl and sulfhydryl). In the antioxidant defense, there was an increase in GSH and SOD in the combination therapy group. Histological analysis showed a reduction in inflammatory infiltrate in the combination therapy group. The number of fibroblasts and the compaction of collagen fibers did not obtain significant responses. Conclusions: Results analyzed together showed that the combined therapy favored the repair process, and that studies can be carried out to enhance the histological analysis therapy favored the tissue repair process and that studies can be carried out to enhance the histological analysis.
Collapse
Affiliation(s)
- Rubya Pereira Zaccaron
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, 97853Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Rusilania Tozi Barbieri
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, 97853Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Carolini Mendes
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, 97853Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Ligia Milanez Venturini
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, 97853Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Naiara Alves
- Graduate Program of Biomedical Science, Herminio Ometto Foundation, Araras-SP, Brazil
| | | | | | | | - Paulo Emílio Feuser
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, 97853Universidade do Extremo Sul Catarinense, Criciúma, Brazil.,Department of Chemical Engineering and Food Engineering, 28117Federal University of Santa Catarina, Florianopolis, Brazil
| | - Anand Thirupathi
- Faculty of Sports Science, 47862Ningbo University, Ningbo, China
| | - Ricardo Andrez Machado-de-Ávila
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, 97853Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Phisiopatology, Program of postgraduate in Science of Health, 97853Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| |
Collapse
|
25
|
Hayun Y, Yaacobi DS, Shachar T, Harats M, Grush AE, Olshinka A. Novel Technologies in Chronic Wound Care. Semin Plast Surg 2022; 36:75-82. [DOI: 10.1055/s-0042-1749095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
AbstractIn Israel, 20% of wounds do not progress to full healing under treatment with conservative technologies of which 1 to 2% are eventually defined as chronic wounds. Chronic wounds are a complex health burden for patients and pose considerable therapeutic and budgetary burden on health systems. The causes of chronic wounds include systemic and local factors. Initial treatment involves the usual therapeutic means, but as healing does not progress, more advanced therapeutic technologies are used. Undoubtedly, advanced means, such as negative pressure systems, and advanced technologies, such as oxygen systems and micrografts, have vastly improved the treatment of chronic wounds. Our service specializes in treating ulcers and difficult-to-heal wounds while providing a multiprofessional medical response. Herein, we present our experience and protocols in treating chronic wounds using a variety of advanced dressings and technologies.
Collapse
Affiliation(s)
- Yehiel Hayun
- Department of Plastic Surgery and Burns, Rabin Medical Center—Beilinson Hospital, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dafna Shilo Yaacobi
- Department of Plastic Surgery and Burns, Rabin Medical Center—Beilinson Hospital, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tal Shachar
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Moti Harats
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The National Burn Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Andrew E. Grush
- Division of Plastic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
- Division of Plastic Surgery, Department of Surgery, Texas Children's Hospital, Houston, Texas
| | - Asaf Olshinka
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Plastic Surgery and Burns Unit, Schneider Children's Medical Center, Petach Tikva, Israel
| |
Collapse
|
26
|
Shams SM, Watari I, Saito E, Ono T. Molar extraction alters gastric mucosa and ghrelin expression in rat stomach: A preliminary study. APOS TRENDS IN ORTHODONTICS 2022. [DOI: 10.25259/apos_133_2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Objectives:
Ghrelin is a key regulator of food intake and is considered a hunger hormone that affects cognition, memory, glucose metabolism, and antidepressant effects. Altered occlusion, such as a loss of molars, has been thought to retard digestive function. However, the association between occlusion and digestive function remains poorly understood. Here, we aimed to explore the effect of bilateral maxillary molar extraction on the gastrointestinal mucosa of growing rats and the expression of ghrelin and its receptor, growth hormone secretagogue receptor (GHSR).
Material and Methods:
Twenty-four male 5-week-old Wistar rats were divided into control (CON) and experimental (EXP) groups (n = 12/group). The rats in the EXP group underwent extraction of the bilateral maxillary first, second, and third molars under general anesthesia. Rats in the CON group underwent a sham operation. All rats in both the CON and EXP groups were fed a powder diet and water ad libitum. The body weight of all rats was monitored throughout the EXP period. Rats in both the CON and EXP groups were euthanized on days 14 and 28, and the stomachs were isolated and subjected to histological analysis. Paraffin serial sections were prepared using a microtome for hematoxylin-eosin and immunohistochemical staining using anti-ghrelin and anti-GHSR antibodies. The distribution and expression of ghrelin-immunopositive and GHSR cells were detected and observed under a light microscope. Data were statistically analyzed using t-tests (P < 0.05).
Results:
There were no significant differences in body weight between the CON and EXP groups throughout the EXP period. Histological analysis showed that the area of the submucosa (ASM), and the number of ghrelinimmunopositive cells were significantly decreased in the EXP group compared with the CON group on day 14. Alternatively, there was no significant difference in the ASM and the number of ghrelin-immunopositive cells between the CON and EXP groups on day 28, whereas the number of ghrelin receptors showed no differences across groups. Furthermore, the number of eosinophilic blood cells significantly increased in the EXP group on days 14 and 28.
Conclusion:
Our findings suggest that bilateral maxillary molar extraction may trigger stomach mucosal changes and alter digestive function through ghrelin expression in rats. This is the first report that occlusal deficiency could alter ghrelin expression in the mucosa of the rat stomach, thus raising concerns about the consequential role of ghrelin.
Collapse
Affiliation(s)
- Shahriar Mohd Shams
- Department of Orthodontic Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan,
| | - Ippei Watari
- Department of Orthodontic Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan,
| | - Eri Saito
- Department of Orthodontic Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan,
| | - Takashi Ono
- Department of Orthodontic Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan,
| |
Collapse
|
27
|
Heydari MB, Ghanbari-Movahed Z, Heydari M, Farzaei MH. In vitro study of the mesenchymal stem cells-conditional media role in skin wound healing process: A systematic review. Int Wound J 2022; 19:2210-2223. [PMID: 35412017 DOI: 10.1111/iwj.13796] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/09/2022] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cell (MSC)-conditioned medium (CM) offers a potential opportunity in the skin wound healing treatment. In this systematic review, an overview of the knowledge on this topic has been provided. A multistep search of the PubMed, Scopus and Science Direct database has been performed to identify papers on MSCs-conditional media used in skin wound healing. Eligibility checks were performed based upon predefined selection criteria. Of the 485 articles initially identified, consequently, only 96 articles apparently related to MSC-conditional media were initially assessed for eligibility. Finally, the 32 articles, strictly regarding the in vitro use of MSCs-conditional media in skin wounds, were analysed. The information analysed highlights the efficacy of MSCs-conditional media on skin wound healing in vitro models. The outcome of this review may be used to guide pre-clinical and clinical studies on the role of MSCs-conditional media in skin wound healing.
Collapse
Affiliation(s)
- Mohammad Bagher Heydari
- Specialist General Surgeon, Taleghani Hospital, Kermanshah University of Medical Sciences (KUMS), Kermanshah, Iran
| | - Zahra Ghanbari-Movahed
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Maryam Heydari
- Department of Pharmacy Zabol University of Medical Sciences, Zabol, Iran
| | - Mohammad Hosein Farzaei
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
28
|
Alloreactivity of Allogeneic Mesenchymal Stem/Stromal Cells and Other Cellular Therapies: A Concise Review. Stem Cells Int 2022; 2022:9589600. [PMID: 35308830 PMCID: PMC8926542 DOI: 10.1155/2022/9589600] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/08/2021] [Accepted: 02/23/2022] [Indexed: 02/08/2023] Open
Abstract
Cellular therapies, deemed live medicine, have brought a wave of new generation biological therapies to treat previously untreatable diseases such as cancers and degenerative diseases like osteoarthritis. These cellular therapies have gained significant recognition in clinical research. The area has been further strengthened with the approval of Chimeric Antigen Receptor added on T cells (CAR-T) therapies by the regulatory authorities USA's Food and Drugs Administration (FDA), European Medical Agency (EMA), the Australian Therapeutic Goods Administration (TGA), and in many countries in 2017 to treat hematological cancers. Another milestone was achieved when allogeneic Mesenchymal Stem Cell- (MSC-) based therapy was approved by the EMA to treat Chrohn's disease in 2018. Allogeneic donor-derived MSC therapies in particular hold great promise and real hope because of their ‘off-the shelf' availability and accessibility for patients in need of urgent treatment. So far, thousands of clinical trials have explored the safety and efficacy of both autologous and allogeneic cell therapies, deeming them safe, however with varying degrees of efficacy. In the current pandemic, clinical trials have begun in many parts of the world to treat severe cases of COVID with MSCs. However, the risk of tissue rejection and the development of undesirable effects due to alloreactivity of allogeneic cells are currently not adequately addressed. Therefore, this warrants careful investigation and detailed reporting of such events by clinical researchers. This review aims at discussing the current landscape of approved allogeneic MSCs along with a few other cellular therapies. We explore any possible reactivity reported to inform the readers of any safety concern and on the efficacy of such therapies.
Collapse
|
29
|
Liu G, ZHOU YUAN, Zhang X, Guo S. Advances in Hydrogels for Stem Cell Therapy: Regulation Mechanisms and Tissue Engineering Applications. J Mater Chem B 2022; 10:5520-5536. [DOI: 10.1039/d2tb01044e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Stem cell therapy has shown unparalleled potential in tissue engineering, but it still faces challenges in the regulation of stem cell fate. Inspired by the native stem cell niche, a...
Collapse
|
30
|
Wu CC, Chen YC, Wu YC, Huang SH, Kuo YR, Lee SS. Foraging for the Optimal Dressing Scaffold to Carry Adipose-Derived Stromal/Progenitor Cells for Cell Therapy. Cell Transplant 2022; 31:9636897221113798. [PMID: 35876233 PMCID: PMC9326840 DOI: 10.1177/09636897221113798] [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] [Indexed: 11/15/2022] Open
Abstract
In our daily plastic surgery practice, we have seen many chronic wounds that need new biotechnology to help and improve wound healing. Stem cells play a crucial role in regenerative medicine. Many pre-clinical researches had reported the beneficial paracrine effects of stem cell therapy for chronic wounds. Cell-friendly scaffolds may provide the protection and three-dimensional space required for adherence of stem cells, thus allowing these stem cells to proliferate and differentiate for treatment purpose. A successful scaffold may enhance the effects of stem cell therapy. In this presented series, the authors attempted to identify the most suitable scaffolds from several commercially available wound dressings that could sustain adipose-derived stromal/progenitor cells (ADSCs) survival. Therefore, we isolated ADSCs containing the green fluorescent protein (GFP) from GFP transgenic rats. The GFP (+) ADSCs and their progenies could be easily observed using a fluorescence microscope. Moreover, we analyzed the cytokines secreted in condition medium (CM) to understand the activities of ADSCs in various dressings. Our results showed that the foam dressings, hydrofiber, chitosan, and alginate plus carboxymethylcellulose were identified as the most suitable dressing materials. Higher concentrations of transforming growth factor beta (TGF-β) and vascular endothelial growth factor (VEGF) were observed 48 h after loading them with GFP (+) ADSCs. Therefore, multiple topical cell therapy using ADSCs can be performed by applying suitable dressing scaffolds without repeated needle injections to deliver the stem cells into the wound bed. Based on their fluorescence property, the GFP (+) ADSCs can also possibly be used for testing biocompatibility of medical materials in the future.
Collapse
Affiliation(s)
- Chia-Chieh Wu
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Orthopedics & Sports Medicine Laboratory, Changhua Christian Hospital, Changhua, Taiwan.,Orthopedic Surgery Department, Changhua Christian Hospital, Changhua, Taiwan.,Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Ying-Che Chen
- Department of Surgery, Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan
| | - Yi-Chia Wu
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Hung Huang
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yur-Ren Kuo
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Su-Shin Lee
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Surgery, Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan.,Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
31
|
Domaszewska-Szostek A, Gewartowska M, Stanczyk M, Narowska B, Moscicka-Wesołowska M, Olszewski WL. An Anhydrous Sodium Chloride Skin Preservation Model for Studies on Keratinocytes Grafting into the Wounds. Pharmaceutics 2021; 13:pharmaceutics13122078. [PMID: 34959359 PMCID: PMC8705222 DOI: 10.3390/pharmaceutics13122078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 01/13/2023] Open
Abstract
Background. Human skin is needed for covering large body areas lost by trauma. The shortcomings of contemporary methods of skin storage are limited preservation time and high immunogenicity if allogeneic. Methods. We investigated whether long-lasting skin preservation in anhydrous sodium chloride (NaCl) may be the source of keratinocytes (KCs) for transplantation. Dehydrated skin fragments were preserved for a time frame from 1 week to 12 months. Then, skin fragments were rehydrated, and KCs were isolated. The viability of KCs was assessed in viability/cytotoxicity test. NaCl-preserved KCs were cultured for 7 days and transplanted to the dorsum of SCID mice. Results. The morphology of NaCl-preserved KCs was unaltered. KCs from all epidermal layers could be identified. All grafts were accepted by the recipients. Transplanted KCs: synthesized keratins 10 and 16 expressed antigens specific for stem cells and transient-amplifying cells, and remained HLA-I-positive. Moreover, they expressed the proliferative marker PCNA. Cells isolated from transplants remained viable and produced enzymes. Conclusions. Transplantation of KCs obtained from human skin and stored in anhydrous NaCl may be considered for the closure of extensive skin wounds. The originality of this method consists of an effective storage procedure and easy preparation of keratinocytes for transplantation.
Collapse
Affiliation(s)
- Anna Domaszewska-Szostek
- Department of Human Epigenetics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Correspondence:
| | - Magdalena Gewartowska
- Electron Microscopy Research Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Marek Stanczyk
- Faculty of Medicine, Lazarski University, 02-662 Warsaw, Poland;
| | - Beata Narowska
- Department of Advanced Material Technologies, Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland;
| | - Maria Moscicka-Wesołowska
- Department of Surgical Research and Transplantology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (M.M.-W.); (W.L.O.)
| | - Waldemar Lech Olszewski
- Department of Surgical Research and Transplantology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (M.M.-W.); (W.L.O.)
| |
Collapse
|
32
|
Sivaraj D, Chen K, Chattopadhyay A, Henn D, Wu W, Noishiki C, Magbual NJ, Mittal S, Mermin-Bunnell AM, Bonham CA, Trotsyuk AA, Barrera JA, Padmanabhan J, Januszyk M, Gurtner GC. Hydrogel Scaffolds to Deliver Cell Therapies for Wound Healing. Front Bioeng Biotechnol 2021; 9:660145. [PMID: 34012956 PMCID: PMC8126987 DOI: 10.3389/fbioe.2021.660145] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Cutaneous wounds are a growing global health burden as a result of an aging population coupled with increasing incidence of diabetes, obesity, and cancer. Cell-based approaches have been used to treat wounds due to their secretory, immunomodulatory, and regenerative effects, and recent studies have highlighted that delivery of stem cells may provide the most benefits. Delivering these cells to wounds with direct injection has been associated with low viability, transient retention, and overall poor efficacy. The use of bioactive scaffolds provides a promising method to improve cell therapy delivery. Specifically, hydrogels provide a physiologic microenvironment for transplanted cells, including mechanical support and protection from native immune cells, and cell-hydrogel interactions may be tailored based on specific tissue properties. In this review, we describe the current and future directions of various cell therapies and usage of hydrogels to deliver these cells for wound healing applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Geoffrey C. Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
33
|
Cannula Size Effect on Stromal Vascular Fraction Content of Fat Grafts. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2021; 9:e3471. [PMID: 33907655 PMCID: PMC8062151 DOI: 10.1097/gox.0000000000003471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/07/2021] [Indexed: 12/13/2022]
Abstract
Background Fat is an active and dynamic tissue composed of adipocytes supported by a structural framework known as the stromal vascular fraction (SVF). SVF is traditionally isolated by enzymatic processing, but new methods are being investigated to isolate it mechanically. Recent studies propose that fat harvested with larger cannulas has a higher survival rate, most likely due to a higher concentration of SVF. Methods Lipoaspirates were obtained from 10 patients who underwent elective liposuction using a 5-mm and a 1-mm cannula attached to a syringe using standard pressure. The fat was aspirated from the same area at adjacent sites. An estimated 5-mm fat particles were also cut down to 1-mm using a micronizer (Marina Medical). A 5-cm3 volume of each sample was compressed through a 0.5-mm opening strainer and rinsed with normal saline to extrude the oil. The resultant SVF left on the strainer was then measured in a 1-cm3 syringe. Results The volume extracted from a 5-mm cannula (mean, 0.23 cm3; SD, 0.10) versus a 1-mm cannula (mean, 0.11 cm3; SD, 0.06) was statistically significant (P = 0.009). An H&E-stained slide from the SVF was obtained for confirmation. Finally, 5-mm fat particles cut down to 1-mm particles using the micronizer resulted in an average volume of 0.20 cm3, which was higher than the average volume harvested with a 1-mm cannula. Conclusions Harvesting with a 5-mm cannula resulted in significantly more SVF than harvesting with a 1-mm cannula. Resizing fat particles harvested with a larger cannula down to 1-mm resulted in higher SVF than SVF obtained with a 1-mm cannula directly.
Collapse
|
34
|
Stromal-vascular fraction of adipose tissue as an alternative source of cellular material for regenerative medicine. UKRAINIAN BIOCHEMICAL JOURNAL 2021. [DOI: 10.15407/ubj93.01.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Adipose tissue is the most convenient source of cellular material for regenerative medicine as it can be obtained in significant quantities via cosmetic liposuction, lipoaspiration of subcutaneous fat or by excision of fat deposits. Adipose tissue consists of adipocytes and cells, which are the part of the stromal-vascular fraction (SVF). Different cell populations can be isolated from SVF, among which the population of adipose tissue stem cells (adipose-derived stem cells, ADSC) is especially important for regenerative medicine. SVF can be obtained relatively easily from adipose tissue (adipose tissue is an alternative to bone marrow in terms of being a source of stem cells) and used to treat various pathologies. Recent studies show that SVF not only has a therapeutic effect similar to that of ADSC, but in some cases is even more effective. The article provides the analysis of the main methods of SVF obtainment, characteristics of SVF cellular composition, its potential for use in clinical medicine and its main advantages over other sources of cellular material, including ADSC cultured in vitro, for regenerative medicine. Keywords: adipocytes, adipose-derived stem cells, regenerative medicine, stromal-vascular fraction
Collapse
|
35
|
Zhang K, Liu F, Zhang Y, Huang X, Tang M, Hou Y, Lv Q, Jin D, Li Y, Kong L. Mechanical Vibration-Extracted Stromal Vascular Fraction Improves Volume Retention after Autologous Fat Grafting. Plast Reconstr Surg 2020; 146:1275-1284. [PMID: 33234957 DOI: 10.1097/prs.0000000000007341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND The stromal vascular fraction can improve volume retention after fat grafting, but the optimal stromal vascular fraction extraction method remains controversial. This study investigated the effect of mechanical vibration on stromal vascular fraction activity and explored the efficacy of vibration as a new extraction method compared to centrifugation, enzyme digestion, and nanoemulsion methods. METHODS Twenty-four rabbits were divided into three groups, and adipose tissue was harvested from the scapular region of each rabbit. In the first group, stromal vascular fraction was extracted from adipose tissue by vibration with different frequencies and durations. Cell counts and colony formation were assessed to determine the optimal vibration parameters. In the second group, stromal vascular fraction was extracted by the four methods, and the cell counts, proliferation, and adipogenic capabilities were observed in vitro. In the third group, adipose tissue mixed with stromal vascular fraction extracted by means of the four methods was grafted into rabbit ears. Volume retention and histologic changes were evaluated over 24 weeks. RESULTS Stromal vascular fraction activity was not influenced by low-frequency (≤45 Hz) and short-duration (≤20 minutes) vibrations. Vibration at 30 Hz for 15 minutes was most efficient for stromal vascular fraction extraction. In vitro, stromal vascular fraction extracted by vibration showed advantages for cell viability. In vivo, the vibration group showed a more normal tissue morphology and a higher retention rate (60.68 ± 7.07 percent) than the enzyme digestion (31.88 ± 4.99 percent), centrifugation (43.76 ± 4.32 percent), and nanoemulsion groups (21.79 ± 3.57 percent) (p < 0.05). CONCLUSION Vibration at 30 Hz for 15 minutes is recommended as a novel nonenzymatic method to extract stromal vascular fraction with high activity.
Collapse
Affiliation(s)
- Kai Zhang
- From the State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases; and the Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University
| | - Fuwei Liu
- From the State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases; and the Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University
| | - Yanyuan Zhang
- From the State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases; and the Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University
| | - Xin Huang
- From the State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases; and the Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University
| | - Mingyue Tang
- From the State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases; and the Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University
| | - Yan Hou
- From the State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases; and the Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University
| | - Qianxin Lv
- From the State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases; and the Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University
| | - Dan Jin
- From the State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases; and the Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University
| | - Yunpeng Li
- From the State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases; and the Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University
| | - Liang Kong
- From the State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases; and the Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University
| |
Collapse
|
36
|
Biostimulative effect of laser on growth of mesenchymal stem/stromal cells in vitro. Postepy Dermatol Alergol 2020; 37:771-780. [PMID: 33240019 PMCID: PMC7675089 DOI: 10.5114/ada.2020.100487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/14/2020] [Indexed: 12/23/2022] Open
Abstract
Introduction Human adipose tissue-derived mesenchymal stem/stromal cells (hAT-MSCs) are multipotent stromal cells with a high potential application in tissue engineering and regenerative medicine. Laser irradiation of the place where the cells were implanted can stimulate their proliferation, increase the secretion of growth factors and thus increase the therapeutic effect. Aim To evaluate the influence of two lasers: Er:YAG and diode on the growth of hAT-MSCs in vitro. Material and methods hAT-MSCs were isolated from human subcutaneous adipose tissue. Immunophenotype of hAT-MSCs was confirmed by flow cytometry. Multipotency of hAT-MSCs was confirmed by differentiation into adipogenic, osteogenic and chondrogenic lineages. hAT-MSCs were irradiated with Er:YAG laser (wavelength 2940 nm, frequency 5, 10 Hz, doses: 0.1–1.2 J/cm2) for 2 s and 4 s and diode laser (wavelength 635 nm and doses: 1–8 J/cm2) for 5, 10, 20, 30 and 40 s. Cell viability was analysed 24 h after the exposure using MTT assay. Results Growth stimulation of hAT-MSCs after 5 Hz Er:YAG laser exposure, 0.1 J/cm2 dose for 4 s and 0.3 J/cm2 dose for 4 s was shown in comparison with the control group. Significant growth stimulation of hAT-MSCs after diode laser irradiation in doses of 1–4 J/cm2 was demonstrated compared to the control group. Conclusions The presented results indicate that both lasers, Er:YAG and diode can be used to stimulate stem/stromal cell growth in vitro. The biostimulative effect of laser therapy on stromal cells may be used in the future in aesthetic dermatology in combined laser and cell therapy.
Collapse
|
37
|
Abstract
INTRODUCTION Transplantation of the keratinocytes, fibroblasts, bone marrow, and adipose tissue-derived mesenchymal stem cells may improve chronic wound healing by delivery of different cytokines, chemokines, and growth factors, which play an essential role in wound healing. The purposes of this review were to check which cell lines are potentially beneficial in enhancement of wound healing and to describe the safety and efficacy of cell therapies in the clinical treatment of chronic wounds, as well as to summarize the pertinent literature and research progress in this field. METHODS PubMed search engine and ClinicalTrials.gov were used to analyze the available data on cell therapies applied in treatment of chronic wound. The analysis included 51 articles, assessing the use of keratinocytes (10), fibroblasts (7), keratinocytes and fibroblasts (10), bone marrow-derived cells (20), and adipose tissue cells (4). Studies on the cell-based products that are currently available on the market (Dermagraft, EpiDex, Apligraf, and HP802-247) were also included, with majority of reports found on fibroblasts and keratinocytes studies. RESULTS Cell-based therapies have a great potential to improve wound healing without major surgical procedures and donor-site morbidity. There is, however, a lack of guidelines on how the age of the patients, the general health conditions, and the coexistence of different diseases may affect the success of these therapies. Further studies are needed to determine the fate of transplanted cells and the number of cells required to obtain optimal effects and outcomes. CONCLUSIONS Despite many promising clinical trials on application of various stem cell-based therapies for treatment of chronic wounds, there is still a need for multicenter comparative studies assessing the dose response and the cell source response on the efficacy of chronic wound healing.
Collapse
|
38
|
Wang S, Hou Y, Li X, Song Z, Sun B, Li X, Zhang H. Comparison of exosomes derived from induced pluripotent stem cells and mesenchymal stem cells as therapeutic nanoparticles for treatment of corneal epithelial defects. Aging (Albany NY) 2020; 12:19546-19562. [PMID: 33049719 PMCID: PMC7732275 DOI: 10.18632/aging.103904] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023]
Abstract
Induced pluripotent stem cells and mesenchymal stem cells are pluripotent stem cells that represent promising therapies for treating various tissue injuries and wound healing. Exosomes are nanosized extracellular vesicles that have been identified as important mediators of therapeutic functions, which are performed via cell communication. In this study, we compared the efficacy of induced pluripotent stem cells-derived exosomes (iPSCs-Exos) and mesenchymal stem cells-derived exosomes (MSCs-Exos) in treating corneal epithelial defects. The characteristics of the two types of exosomes were not significantly different. Compared to MSCs-Exos, iPSCs-Exos had a better in vitro effect on the proliferation, migration, cell cycle promotion and apoptosis inhibition of human corneal epithelial cells. iPSCs/MSCs-Exos promoted cell regeneration by upregulating cyclin A and CDK2 to drive HCECs to enter the S phase from the G0/G1 phase. In vivo results from a corneal epithelial defect model showed that both iPSCs-Exos and MSCs-Exos accelerated corneal epithelium defect healing while the effects of iPSCs-Exos were much stronger than those of MSCs-Exos. This study demonstrated that iPSCs-Exos had a better therapeutic effect on corneal epithelial defect healing. Thus, a novel potential nanotherapeutic strategy for treating corneal epithelial defects and even more ocular surface disease could be undertaken by using iPSCs-Exos dissolved in eye drops.
Collapse
Affiliation(s)
- Shudan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yunlong Hou
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China,National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Shijiazhuang 050200, China
| | - Xuran Li
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Zhen Song
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Baoqi Sun
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, Weifang 261042, China
| | - Xinyue Li
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Hong Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|
39
|
Shin DY, Park JU, Choi MH, Kim S, Kim HE, Jeong SH. Polydeoxyribonucleotide-delivering therapeutic hydrogel for diabetic wound healing. Sci Rep 2020; 10:16811. [PMID: 33033366 PMCID: PMC7546631 DOI: 10.1038/s41598-020-74004-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
Patients with diabetes experience delayed wound healing because of the uncontrolled glucose level in their bloodstream, which leads to impaired function of white blood cells, poor circulation, decreased production and repair of new blood vessels. Treatment using polydeoxyribonucleotide (PDRN), which is a DNA extracted from the sperm cells of salmon, has been introduced to accelerate the healing process of diabetic wounds. To accelerate the wound-healing process, sustained delivery of PDRN is critical. In this study, taking advantage of the non-invasive gelation property of alginate, PDRN was loaded inside the hydrogel (Alg-PDRN). The release behavior of PDRN was altered by controlling the crosslinking density of the Alg hydrogel. The amount of PDRN was the greatest inside the hydrogel with the highest crosslinking density because of the decreased diffusion. However, there was an optimal degree of crosslinking for the effective release of PDRN. In vitro studies using human dermal fibroblasts and diabetes mellitus fibroblasts and an in ovo chorioallantoic membrane assay confirmed that the Alg-PDRN hydrogel effectively induced cell proliferation and expression of angiogenic growth factors and promoted new blood vessel formation. Its effectiveness for accelerated diabetic wound healing was also confirmed in an in-vivo animal experiment using a diabetic mouse model.
Collapse
Affiliation(s)
- Da Yong Shin
- Department of Materials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ji-Ung Park
- Department of Plastic and Reconstructive Surgery, Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Min-Ha Choi
- Department of Plastic and Reconstructive Surgery, Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Sukwha Kim
- Medical Big Data Research Center, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Hyoun-Ee Kim
- Department of Materials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Advanced Institutes of Convergence Technology, Seoul National University, Gwanggyo, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16229, Republic of Korea
| | - Seol-Ha Jeong
- Department of Materials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
40
|
Ajayi AF, Olaleye BS. Age-related changes in haematological parameters and biochemical markers of healing in the stomach of rats with acetic acid induced injury. Toxicol Rep 2020; 7:1272-1281. [PMID: 32995300 PMCID: PMC7511975 DOI: 10.1016/j.toxrep.2020.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023] Open
Abstract
This study examined the changes in haematological and biochemical variables in response to gastric mucosa injury in male Wistar rats divided into four groups according to their ages (3, 6, 12, and 18 months). 0.2 ml of acetic acid was injected intraluminal into the stomach glandular portion of each rat for 45 seconds under anaesthesia. Collection of blood and stomach samples occurred on days 3, 7, 14 and 21 post-induction of gastric ulcer. The results obtained from this study showed 100 % area of gastric mucosa healed in 3-month old rats, 91.72 %, 68.52 % and 62.81 % area of mucosa treated in 6, 12 and 18-month old rats respectively on day 21 post-induction of gastric ulcer. Increased circulation of blood cells in younger rats occurred, neutrophil-lymphocyte ratio (NLR) was decreased in younger rats (3 and 6 months) significantly (p < 0.05) when compared to older rats (12 and 18 months). Lipid peroxidation and glutathione (GSH) levels were elevated in older rats (12 and 18 months) significantly (p < 0.05) when compared to younger rats (3 and 6 months). In comparison, superoxide dismutase (SOD) and catalase levels were decreased in older rats (12 and 18 months) significantly (p < 0.05) when compared to younger rats (3 and 6 months). Histological evaluation showed evidence of early healing with re-epithelialisation and angiogenesis in younger rats, but older rats showed delayed healing. The study showed that the slower rate of healing of gastric ulcer with advancing age in rats might be due to reducing circulating blood cells and anti-inflammatory activities during healing via a lipid peroxidation-dependent mechanism.
Collapse
Key Words
- ALP, Alkaline phosphatase
- ALT, Alanine aminotransferase
- ANOVA, Analysis of variance
- AST, Aspartate aminotransferase
- Age
- Angiogenesis
- DTNB, 5, 5' –Dithiobis-2-nitrobenzoic acid
- EGF, Epithelial growth factor
- GSH, Glutathione
- HB, Haemoglobin
- Haematology
- KIM-1, Kidney injury molecule-1
- Lipid peroxidation
- MDA, Malondialdehyde
- MDA-TBA, Malondialdehyde-thiobarbituric acid
- NLR, Neutrophil-lymphocyte ratio
- NSAID, Nonsteroidal anti-inflammatory drugs
- Neutrophil-lymphocyte ratio
- PDEGF, Platelet-derived endothelial growth factor
- PLT, Platelets
- RBC, Red blood cell
- ROS, Reactive oxygen species
- Re-epithelialisation
- SOD, Superoxide dismutase
- TFF, 3 Trefoil factor 3
- VEGF, vascular endothelial growth factor
- WBC, White blood cell
Collapse
Affiliation(s)
- Ayodeji F Ajayi
- Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria.,Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Babafemi S Olaleye
- Department of Physiology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
41
|
Nilforoushzadeh MA, Khodadadi Yazdi M, Baradaran Ghavami S, Farokhimanesh S, Mohammadi Amirabad L, Zarrintaj P, Saeb MR, Hamblin MR, Zare M, Mozafari M. Mesenchymal Stem Cell Spheroids Embedded in an Injectable Thermosensitive Hydrogel: An In Situ Drug Formation Platform for Accelerated Wound Healing. ACS Biomater Sci Eng 2020; 6:5096-5109. [PMID: 33455261 DOI: 10.1021/acsbiomaterials.0c00988] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The ability of mesenchymal stem cells (MSCs) to enhance cutaneous wound healing has been well established. Extensive expansion of cells to reach sufficient cell numbers for regenerating tissues has always limited cell-based therapies. An ingenious solution to address this challenge is to develop a strategy to increase the immunomodulatory effects of MSCs without expanding them. In this study, we employed a simple characteristic of cells. It was observed that an optimized three-dimensional (3D) MSC culture in spheroid forms significantly improved their paracrine effects. An electrospray (ES) encapsulation apparatus was used to encapsulate individual or 3D spheroid MSCs into microscale alginate beads (microbeads). Furthermore, alginate microbeads were embedded in an injectable thermosensitive hydrogel matrix, which gels at skin temperature. The hydrogel fills and seals the wounds cavities, maintains high humidity at the wound area, absorbs exudate, and fixes microbeads, protecting them from direct contact with the harsh wound environment. In vitro investigations revealed that secretion of interleukin 10 (IL-0) and transforming growth factor β1 (TGF-β1) gene was gradually enhanced, providing a delivery platform for prolonged release of bioactive molecules. In vivo study on full-thickness wounds showed granulation and re-epithelialization, only after 7 days. Moreover, increased expression of α-smooth muscle actin (α-SMA) in the first 14 days after treatment ensured wound contraction. Besides, a gradual decrease in α-SMA secretion resulted in reduced scar formation. Well-organized collagen fibrils and high expression of the angiogenesis biomarker CD31 confirmed the promoting effect of the hydrogel on the wound-healing process. The proposed wound-dressing system would potentially be used in scalable and effective cell-based wound therapies.
Collapse
Affiliation(s)
| | | | - Shaghayegh Baradaran Ghavami
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samila Farokhimanesh
- Department of Biotechnology, Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Payam Zarrintaj
- School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, Oklahoma 74078, United States
| | - Mohammad Reza Saeb
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Department of Dermatology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Mehrak Zare
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Potential of Tissue-Engineered and Artificial Dermis Grafts for Fingertip Reconstruction. Plast Reconstr Surg 2020; 146:1082-1095. [PMID: 32915527 DOI: 10.1097/prs.0000000000007258] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Management of skin and soft-tissue defects of the fingertips is functionally and aesthetically important, but controversial, especially when bones are exposed. Recent advances in wound healing technology allow the use of cells or biological dermis. The authors studied the clinical efficacy of tissue-engineered dermis grafts and artificial dermis grafts versus immediate reconstructive procedures, such as the reverse digital artery island flap, in treating bone-exposed fingertip defects. METHODS One hundred eighty-two patients with bone-exposed fingertip defects treated with tissue-engineered dermis grafts (n = 71), artificial dermis grafts (n = 23), or reverse digital artery island flaps (n = 88) were included in this retrospective cohort study. Surgical time, duration of hospitalization, total cost, success rate, healing time, sensory recovery, range of motion, scar quality, and patient satisfaction were compared. RESULTS No tissue-engineered or artificial dermis graft exhibited graft rejection or failure, whereas there was one partial loss and one total loss after reverse digital artery island flap surgery. Tissue-engineered dermis grafts were superior in scar quality, and artificial dermis grafts had shorter surgical times and lower surgical costs; both groups demonstrated superior results in postoperative range of motion and sensory recovery in two-point discrimination tests and shorter hospitalization, compared with the reverse digital artery island flap group. The reverse digital artery island flap had shorter complete closure time and less postoperative tingling sensation. There were no differences in overall patient satisfaction among the groups. CONCLUSIONS Tissue-engineered and artificial dermis grafts may be promising alternatives for fingertip reconstruction. In particular, tissue-engineered dermis grafts may deliver superior functional results, including recovery of sensory discomfort and aesthetic results in terms of scar quality over artificial dermis grafts. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, III.
Collapse
|
43
|
Liubaviciute A, Ivaskiene T, Biziuleviciene G. Modulated mesenchymal stromal cells improve skin wound healing. Biologicals 2020; 67:1-8. [DOI: 10.1016/j.biologicals.2020.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/26/2020] [Accepted: 08/04/2020] [Indexed: 12/20/2022] Open
|
44
|
Wang X, Deng M, Yu Z, Cai Y, Liu W, Zhou G, Wang X, Cao Y, Li W, Zhang W. Cell-free fat extract accelerates diabetic wound healing in db/db mice. Am J Transl Res 2020; 12:4216-4227. [PMID: 32913499 PMCID: PMC7476113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/16/2020] [Indexed: 06/11/2023]
Abstract
Cell-free fat extract (CEFFE), the liquid fraction derived from fat tissues, is enriched with a variety of growth factors and possesses pro-angiogenic, anti-apoptotic, and anti-oxidative properties. The aim of this study was to determine if CEFFE could accelerate chronic wound healing in mice with diabetes and investigate its underlying mechanisms. A model of circular full-thickness wound (6 mm diameter) was produced in the central dorsal region of spontaneous type 2 diabetes mellitus db/db mice. The mice were divided to three groups depending on dosage of CEFFE administered for the study; high dose CEFFE group (CEFFEhigh; administered 2.5 ml/kg/day via subcutaneous injection for six days), low dose CEFFE group (CEFFElow; administered 2.5 ml/kg/day via subcutaneous injection for three days), and a control group receiving phosphate buffer solution. Wound closure was evaluated on day 3, 7, 10, and 14 post-operation. Histological analyses, including hematoxylin-eosin staining and Masson's trichrome staining and immunohistological staining of anti-CD31 and anti-CD68, were also performed. Moreover, the effects of CEFFE on proliferation, migration, and tube formation of human immortal keratinocyte cells (HaCaT) and human vascular endothelial cells (HUVEC) were tested in vitro. The results showed that the local injection of CEFFE significantly accelerated wound healing in mice with diabetes. CEFFE improved re-epithelization and collagen secretion, promoted angiogenesis, and inhibited inflammatory macrophage infiltration in vivo. CEFFE also promoted HaCaT proliferation and migration and enhanced tubular formation in cultured HUVEC. It was concluded that CEFFE accelerates wound healing through pro-angiogenic and anti-inflammatory activities.
Collapse
Affiliation(s)
- Xiangsheng Wang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
| | - Mingwu Deng
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
| | - Ziyou Yu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
| | - Yizuo Cai
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
- National Tissue Engineering Center of ChinaShanghai 200041, China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
- National Tissue Engineering Center of ChinaShanghai 200041, China
| | - Xiansong Wang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
| | - Yilin Cao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
- National Tissue Engineering Center of ChinaShanghai 200041, China
| | - Wei Li
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue EngineeringShanghai 200011, China
- National Tissue Engineering Center of ChinaShanghai 200041, China
| |
Collapse
|
45
|
Álvaro-Afonso FJ, Sanz-Corbalán I, Lázaro-Martínez JL, Kakagia D, Papanas N. Adipose-Derived Mesenchymal Stem Cells in the Treatment of Diabetic Foot Ulcers: A Review of Preclinical and Clinical Studies. Angiology 2020; 71:853-863. [PMID: 32723090 DOI: 10.1177/0003319720939467] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This review provides an outline of the use of adipose-derived mesenchymal stem cells (AMSCs) in the treatment of diabetic foot ulcers (DFUs). A systematic search of PubMed and the Cochrane database was performed on October 2, 2019. Eighteen studies were identified (14 preclinical and 4 clinical). Studies in animal models have demonstrated that AMSCs enhance diabetic wound healing, accelerate granulation tissue formation, and increase reepithelialization and neovascularization. Only 1 randomized control trial has been published so far. Patients (n = 25) with DFUs were treated using an allogeneic AMSC directly on the wound bed as a primary dressing, and improvements were found in complete wound closure in the treatment group (n = 16). Three clinical studies showed that autologous AMSC might be a safe alternative to achieve therapeutic angiogenesis in patients with diabetes and peripheral arterial disease. Based on the available evidence, AMSCs hold promise in the treatment of DFUs. However, this evidence requires confirmation by well-designed trials. Additional studies are also required to understand some issues regarding this treatment for DFUs. For example, the potential application of autologous or allogeneic AMSCs in different types of DFUs, optimal dose/infusion schedules, safety evaluations, and cost-effectiveness.
Collapse
Affiliation(s)
- Francisco Javier Álvaro-Afonso
- Diabetic Foot Unit, University Podiatric Clinic, Edificio Facultad de Medicina, 16734Complutense University of Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Irene Sanz-Corbalán
- Diabetic Foot Unit, University Podiatric Clinic, Edificio Facultad de Medicina, 16734Complutense University of Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - José Luis Lázaro-Martínez
- Diabetic Foot Unit, University Podiatric Clinic, Edificio Facultad de Medicina, 16734Complutense University of Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Despoina Kakagia
- Department of Plastic Surgery, Democritus University of Thrace, University Hospital of Alexandroupolis, Greece
| | - Nikolaos Papanas
- Diabetes Centre-Diabetic Foot Clinic, Second Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Greece
| |
Collapse
|
46
|
Raghuram AC, Yu RP, Lo AY, Sung CJ, Bircan M, Thompson HJ, Wong AK. Role of stem cell therapies in treating chronic wounds: A systematic review. World J Stem Cells 2020; 12:659-675. [PMID: 32843920 PMCID: PMC7415243 DOI: 10.4252/wjsc.v12.i7.659] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/03/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The impairment of cutaneous wound healing results in chronic, non-healing wounds that are caused by altered wound environment oxygenation, tissue injury, and permissive microbial growth. Current modalities for the treatment of these wounds inadequately address the complex changes involved in chronic wound pathogenesis. Consequently, stem cell therapies have emerged as a potential therapeutic modality to promote cutaneous regeneration through trophic and paracrine activity.
AIM To investigate current literature regarding use of stem cell therapies for the clinical treatment of chronic, non-healing wounds.
METHODS PubMed, EMBASE, Cochrane Library, Web of Science, and Scopus were queried with combinations of the search terms “mesenchymal stem cells,” “adult stem cells,” “embryonic stem cells,” “erythroid precursor cells,” “stem cell therapies,” and “chronic wounds” in order to find relevant articles published between the years of 2000 and 2019 to review a 20-year experience. Reference lists from the articles were reviewed to identify additional pertinent articles. Retrieved manuscripts (reviews, case reports/series, retrospective/prospective studies, and clinical trials) were evaluated by the authors for their depiction of clinical stem cell therapy use. Data were extracted from the articles using a standardized collection tool.
RESULTS A total of 43 articles describing the use of stem cell therapies for the treatment of chronic wounds were included in this review. While stem cell therapies have been explored in in vitro and in vivo applications in the past, recent efforts are geared towards assessing their clinical role. A review of the literature revealed that adipose-derived stem cells, bone marrow-derived stem cells, bone marrow-derived mononuclear cells, epidermally-derived mesenchymal stem cells, fibroblast stem cells, keratinocyte stem cells, placental mesenchymal stem cells, and umbilical cord mesenchymal stem cells have all been employed in the treatment of chronic wounds of various etiologies. Most recently, embryonic stem cells have emerged as a novel stem cell therapy with the capacity for multifaceted germ cell layer differentiation. With the capacity for self-renewal and differentiation, stem cells can enrich existing cell populations in chronic wounds in order to overcome barriers impeding the progression of wound healing. Further, stem cell therapies can be utilized to augment cell engraftment, signaling and activity, and resultant patient outcomes.
CONCLUSION Assessing observed clinical outcomes, potential for stem cell use, and relevant therapeutic challenges allows wound care stakeholders to make informed decisions regarding optimal treatment approaches for their patients’ chronic wounds.
Collapse
Affiliation(s)
- Anjali C Raghuram
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, United States
| | - Roy P Yu
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, United States
| | - Andrea Y Lo
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, United States
| | - Cynthia J Sung
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, United States
| | - Melissa Bircan
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, United States
| | - Holly J Thompson
- Wilson Dental Library, Herman Ostrow School of Dentistry of USC, Los Angeles, CA 90089, United States
| | - Alex K Wong
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, United States
| |
Collapse
|
47
|
Ha SS, Song ES, Du P, Suhaeri M, Lee JH, Park K. Novel ECM Patch Combines Poly(vinyl alcohol), Human Fibroblast-Derived Matrix, and Mesenchymal Stem Cells for Advanced Wound Healing. ACS Biomater Sci Eng 2020; 6:4266-4275. [PMID: 33463354 DOI: 10.1021/acsbiomaterials.0c00657] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Decellularized extracellular matrix (ECM)-based scaffold has been a very useful resource for effective tissue regeneration. In this study, we report a novel ECM patch that physically combines human fibroblast-derived matrix (hFDM) and poly(vinyl alcohol) (PVA) hydrogel. hFDM was obtained after decellularization of in vitro cultured human fibroblasts. We investigated the basic characteristics of hFDM alone using immunofluorescence (fibronectin, collagen type I) and angiogenesis-related factor analysis. Successful incorporation of hFDM with PVA produced an hFDM/PVA patch, which showed excellent cytocompatibility with human mesenchymal stem cells (hMSCs), as assessed via cell adhesion, viability, and proliferation. Moreover, in vitro scratch assay using human dermal fibroblasts showed a significant improvement of cell migration when treated with the paracrine factors originated from the hMSC-incorporated hFDM. To evaluate the therapeutic effect on wound healing, hMSCs were seeded on the hFDM/PVA patch and they were then transplanted into a mouse full-thickness wound model. Among four experimental groups (control, PVA, hFDM/PVA, hMSC/hFDM/PVA), we found that hMSC/hFDM/PVA patch accelerated the wound closure with time. More notably, histology and immunofluorescence demonstrated that compared to the other interventions tested, hMSC/hFDM/PVA patch could lead to significantly advanced tissue regeneration, as confirmed via nearly normal epidermis thickness, skin adnexa regeneration (hair follicle), mature collagen deposition, and neovascularization. Additionally, cell tracking of prelabeled hMSCs suggests the in vivo retention of transplanted cells in the wound region after the transplantation of hMSC/hFDM/PVA patch. Taken together, our engineered ECM patch supports a strong regenerative potential toward advanced wound healing.
Collapse
Affiliation(s)
- Sang Su Ha
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Eui Sun Song
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Ping Du
- Center for Human Tissues & Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Muhammad Suhaeri
- Unit of Education, Research, and Training, Universitas Indonesia Hospital, Universitas Indonesia, Depok 16424, Indonesia
| | - Jong Ho Lee
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Republic of Korea
| |
Collapse
|
48
|
Rasheed AI, Shawki SR, Mostafa RG, Sharara MA. A comparative clinical and histopathological pilot study of different fractional CO 2 laser parameters in treatment of atrophic linear scars. J Cosmet Dermatol 2020; 20:116-123. [PMID: 32416611 DOI: 10.1111/jocd.13489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 04/22/2020] [Accepted: 05/07/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Atrophic scars cause significant patient morbidity. Fractional photothermolysis is one of the most effective treatment options used to resurface scars of different etiologies. AIMS To assess the efficacy and safety of different fractional ablative CO2 laser parameters in treatment of linear atrophic depressed post-traumatic facial scars in adult male patients. METHODS A prospective pilot study of 20 adult male patients (skin types Π- Ⅳ, aged 18-45) with post-traumatic atrophic linear scars were divided into 2 groups each comprising 10 patients receiving different fractional CO2 laser parameters. Both groups received 3 laser sessions, one month apart, and were followed for 2 months after the last treatment session. Clinical and histological assessments were done to all patients before treatment and 2 months after the last treatment session. Also, patient satisfaction and side effects were recorded. RESULTS The study showed statistically significant reduction in average scar volume in both groups (P < .01), with reduction in depth more obvious than reduction in width or length in both groups. There was a highly significant difference in overall scar improvement represented by scar volume between both groups (P < .01) with an average reduction in scar volume of 42.85% in group (a) compared with 35.29% in group (b). Also, there was a highly statistically significant increase in both epidermal and papillary and reticular dermis thickness in both groups after treatment. However, the difference between both groups was nonsignificant. Side effects were mild, well tolerated, and transient. CONCLUSION Fractional CO2 laser can be utilized as a safe and effective modality in treatment of post-traumatic linear atrophic scars of the face. Adjusting parameters toward increasing depth of penetration and decreasing thermal coagulative effect gives better results.
Collapse
Affiliation(s)
- Ahmad I Rasheed
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Rabab G Mostafa
- Department of Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Manal A Sharara
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
49
|
Huayllani MT, Sarabia-Estrada R, Restrepo DJ, Boczar D, Sisti A, Nguyen JH, Rinker BD, Moran SL, Quiñones-Hinojosa A, Forte AJ. Adipose-derived stem cells in wound healing of full-thickness skin defects: a review of the literature . J Plast Surg Hand Surg 2020; 54:263-279. [PMID: 32427016 DOI: 10.1080/2000656x.2020.1767116] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The complex process of wound healing can be delayed in circumstances when the natural niche is extremely altered. Adipose-derived stem cells (ADSC) seem to be a promising therapy for these type of wounds. We aim to describe the studies that used ADSC for wound healing after a full-thickness skin defect, the ADSC mechanisms of action, and the outcomes of the different ADSC therapies applied to date. We performed a review by querying PubMed database for studies that evaluated the use of ADSC for wound healing. The Mesh terms, adipose stem cells AND (skin injury OR wound healing) and synonyms were used for the search. Our search recorded 312 articles. A total of 30 articles met the inclusion criteria. All were experimental in nature. ADSC was applied directly (5 [16.7%]), in sheets (2 [6.7%]), scaffolds (14 [46.7%]), skin grafts (3 [10%]), skin flaps (1 [3.3%]), as microvesicles or exosomes (4 [13.3%]), with adhesives for wound closure (1 [3.3%]), and in a concentrated conditioned hypoxia-preconditioned medium (1 [3.3%]). Most of the studies reported a benefit of ADSC and improvement of wound healing with all types of ADSC therapy. ADSC applied along with extracellular matrix, stromal cell-derived factor (SDF-1) or keratinocytes, or ADSC seeded in scaffolds showed better outcomes in wound healing than ADSC alone. ADSC have shown to promote angiogenesis, fibroblast migration, and up-regulation of macrophages chemotaxis to enhance the wound healing process. Further studies should be conducted to assure the efficacy and safety of the different ADSC therapies.
Collapse
Affiliation(s)
| | | | | | - Daniel Boczar
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Andrea Sisti
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Justin H Nguyen
- Department of Transplantation Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Brian D Rinker
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Steven L Moran
- Division of Plastic Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Antonio J Forte
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
50
|
Yin S, Yang X, Bi H, Zhao Z. Combined Use of Autologous Stromal Vascular Fraction Cells and Platelet-Rich Plasma for Chronic Ulceration of the Diabetic Lower Limb Improves Wound Healing. INT J LOW EXTR WOUND 2020; 20:135-142. [PMID: 32131655 DOI: 10.1177/1534734620907978] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The discovery of stromal vascular fraction cells and platelet-rich plasma in promoting tissue regeneration has prompted a new idea for the treatment of chronic diabetic ulcer of the lower limb. The study aim was to evaluate the clinical efficacy of a new method that applied stromal vascular fraction cells and platelet-rich plasma together in the treatment of recalcitrant chronic diabetic ulcer. We conducted a single-center, prospective, open, noncontrolled study. Four patients (5 ulcers in total) who had received standard treatment for diabetic ulcer for at least 3 months that failed to heal was enrolled. All patients were treated with surgical debridement, cell suspension (stromal vascular fraction cells suspended by platelet-rich plasma) injection into the wound, and platelet-rich plasma gel coverage. Wounds were measured every week after treatment using a 2-dimensional digital camera and a 3-dimensional wound measurement device. All patients were followed-up for 4 months after the treatment. Four of the 5 ulcers healed completely within a mean of 71.75 ± 29.57 days. The average proportion of granulation tissue achieved 100% within 4 weeks for all cases. The wound size decreased to less than half of the original size for all cases 4 weeks after the treatment. Findings revealed that the new treatment is efficient to achieve wound healing in patients with recalcitrant chronic diabetic ulcer of lower limb.
Collapse
Affiliation(s)
- Shilu Yin
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Xin Yang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Hongsen Bi
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Zhenmin Zhao
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| |
Collapse
|