1
|
Pinheiro-Machado E, Faas MM, de Haan BJ, Moers C, Smink AM. Culturing Conditions Dictate the Composition and Pathways Enrichment of Human and Rat Perirenal Adipose-Derived Stromal Cells' Secretomes. Stem Cell Rev Rep 2024; 20:1869-1888. [PMID: 38922529 PMCID: PMC11445368 DOI: 10.1007/s12015-024-10748-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2024] [Indexed: 06/27/2024]
Abstract
Understanding the impact of various culturing strategies on the secretome composition of adipose-derived stromal cells (ASC) enhances their therapeutic potential. This study investigated changes in the secretome of perirenal ASC (prASC) under different conditions: normoxia, cytokine exposure, high glucose, hypoxia, and hypoxia with high glucose. Using mass spectrometry and enrichment clustering analysis, we found that normoxia enriched pathways related to extracellular matrix (ECM) organization, platelet degranulation, and insulin-like growth factor (IGF) transport and uptake. Cytokine exposure influenced metabolism, vascular development, and protein processing pathways. High glucose affected the immune system, metabolic processes, and IGF transport and uptake. Hypoxia impacted immune and metabolic processes and protein processing. Combined hypoxia and high glucose influenced the immune system, IGF transport and uptake, and ECM organization. Our findings highlight the potential of manipulating culturing conditions to produce secretomes with distinct protein and functional profiles, tailoring therapeutic strategies accordingly.
Collapse
Affiliation(s)
- Erika Pinheiro-Machado
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1 (EA11), Groningen, 9713 GZ, The Netherlands
| | - Marijke M Faas
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1 (EA11), Groningen, 9713 GZ, The Netherlands
| | - Bart J de Haan
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1 (EA11), Groningen, 9713 GZ, The Netherlands
| | - Cyril Moers
- Department of Surgery - Organ Donation and Transplantation, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Alexandra M Smink
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1 (EA11), Groningen, 9713 GZ, The Netherlands.
| |
Collapse
|
2
|
Xu Y, Liu X, Ahmad MA, Ao Q, Yu Y, Shao D, Yu T. Engineering cell-derived extracellular matrix for peripheral nerve regeneration. Mater Today Bio 2024; 27:101125. [PMID: 38979129 PMCID: PMC11228803 DOI: 10.1016/j.mtbio.2024.101125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/28/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024] Open
Abstract
Extracellular matrices (ECMs) play a key role in nerve repair and are recognized as the natural source of biomaterials. In parallel to extensively studied tissue-derived ECMs (ts-ECMs), cell-derived ECMs (cd-ECMs) also have the capability to partially recapitulate the complicated regenerative microenvironment of native nerve tissues. Notably, cd-ECMs can avoid the shortcomings of ts-ECMs. Cd-ECMs can be prepared by culturing various cells or even autologous cells in vitro under pathogen-free conditions. And mild decellularization can achieve efficient removal of immunogenic components in cd-ECMs. Moreover, cd-ECMs are more readily customizable to achieve the desired functional properties. These advantages have garnered significant attention for the potential of cd-ECMs in neuroregenerative medicine. As promising biomaterials, cd-ECMs bring new hope for the effective treatment of peripheral nerve injuries. Herein, this review comprehensively examines current knowledge about the functional characteristics of cd-ECMs and their mechanisms of interaction with cells in nerve regeneration, with a particular focus on the preparation, engineering optimization, and scalability of cd-ECMs. The applications of cd-ECMs from distinct cell sources reported in peripheral nerve tissue engineering are highlighted and summarized. Furthermore, current limitations that should be addressed and outlooks related to clinical translation are put forward as well.
Collapse
Affiliation(s)
- Yingxi Xu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xianbo Liu
- Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | | | - Qiang Ao
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, China
| | - Yang Yu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, China
| | - Dan Shao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, Guangzhou, China
| | - Tianhao Yu
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| |
Collapse
|
3
|
Ruoss S, Nasamran CA, Ball ST, Chen JL, Halter KN, Bruno KA, Whisenant TC, Parekh JN, Dorn SN, Esparza MC, Bremner SN, Fisch KM, Engler AJ, Ward SR. Comparative single-cell transcriptional and proteomic atlas of clinical-grade injectable mesenchymal source tissues. SCIENCE ADVANCES 2024; 10:eadn2831. [PMID: 38996032 PMCID: PMC11244553 DOI: 10.1126/sciadv.adn2831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 06/06/2024] [Indexed: 07/14/2024]
Abstract
Bone marrow aspirate concentrate (BMAC) and adipose-derived stromal vascular fraction (ADSVF) are the most marketed stem cell therapies to treat a variety of conditions in the general population and elite athletes. Both tissues have been used interchangeably clinically even though their detailed composition, heterogeneity, and mechanisms of action have neither been rigorously inventoried nor compared. This lack of information has prevented investigations into ideal dosages and has facilitated anecdata and misinformation. Here, we analyzed single-cell transcriptomes, proteomes, and flow cytometry profiles from paired clinical-grade BMAC and ADSVF. This comparative transcriptional atlas challenges the prevalent notion that there is one therapeutic cell type present in both tissues. We also provide data of surface markers that may enable isolation and investigation of cell (sub)populations. Furthermore, the proteome atlas highlights intertissue and interpatient heterogeneity of injected proteins with potentially regenerative or immunomodulatory capacities. An interactive webtool is available online.
Collapse
Affiliation(s)
- Severin Ruoss
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | - Chanond A. Nasamran
- Center for Computational Biology and Bioinformatics, UC San Diego, La Jolla, CA, USA
| | - Scott T. Ball
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | - Jeffrey L. Chen
- Department of Anesthesiology, Center for Pain, UC San Diego, La Jolla, CA, USA
| | - Kenneth N. Halter
- Department of Anesthesiology, Center for Pain, UC San Diego, La Jolla, CA, USA
| | - Kelly A. Bruno
- Department of Anesthesiology, Center for Pain, UC San Diego, La Jolla, CA, USA
| | - Thomas C. Whisenant
- Center for Computational Biology and Bioinformatics, UC San Diego, La Jolla, CA, USA
| | - Jesal N. Parekh
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | - Shanelle N. Dorn
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | - Mary C. Esparza
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | | | - Kathleen M. Fisch
- Center for Computational Biology and Bioinformatics, UC San Diego, La Jolla, CA, USA
- Department of Obstetrics, Gynecology & Reproductive Sciences, UC San Diego, La Jolla, CA, USA
| | - Adam J. Engler
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Samuel R. Ward
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA, USA
- Department of Radiology, UC San Diego, La Jolla, CA, USA
| |
Collapse
|
4
|
Cruciani S, Coradduzza D, Balzano F, Garroni G, Azara E, Pala R, Delitala AP, Madonia M, Tedde A, Capobianco G, Petrillo M, Angelucci C, Carru C, Ventura C, Maioli M. Modulation of adipose-derived stem cell behavior by prostate pathology-associated plasma: insights from in vitro exposure. Sci Rep 2024; 14:14765. [PMID: 38926454 PMCID: PMC11208502 DOI: 10.1038/s41598-024-64625-0] [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: 03/16/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Adipose-derived stem cells (ADSCs) are promising in regenerative medicine. Their proliferation, survival and activation are influenced by specific signals within their microenvironment, also known as niche. The stem cell niche is regulated by complex interactions between multiple cell types. When transplanted in a specific area, ADSCs can secrete several immunomodulatory factors. At the same time, a tumor microenvironment can influence stem cell behavior, modulating proliferation and their ability to differentiate into a specific phenotype. Whitin this context, we exposed ADSCs to plasma samples derived from human patients diagnosed with prostate cancer (PC), or precancerous lesions (PL), or benign prostatic hyperplasia (BPH) for 4, 7 or 10 days. We then analyzed the expression of main stemness-related markers and cell-cycle regulators. We also measured cytokine production and polyamine secretion in culture medium and evaluated cell morphology and collagen production by confocal microscopy. The results obtained from this study show significant changes in the morphology of ADSCs exposed to plasma samples, especially in the presence of prostate cancer plasma, suggesting important implications in the use of ADSCs for the development of new treatments and application in regenerative medicine.
Collapse
Affiliation(s)
- Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Donatella Coradduzza
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Francesca Balzano
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Giuseppe Garroni
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Emanuela Azara
- Institute of Biomolecular Chemistry, National Research Council, 07100, Sassari, Italy
| | - Renzo Pala
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Alessandro P Delitala
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Massimo Madonia
- Department of Clinical and Experimental Medicine, Urologic Clinic, University of Sassari, Sassari, Italy
| | - Alessandro Tedde
- Department of Clinical and Experimental Medicine, Urologic Clinic, University of Sassari, Sassari, Italy
| | - Giampiero Capobianco
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Marco Petrillo
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Cecilia Angelucci
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
- Medical Oncology Unit, University Hospital (AOU) of Sassari, 07100, Sassari, Italy
| | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering, Istituto Nazionale Biostrutture E Biosistemi (INBB)-Eldor Lab, Via Corticella 183, 40128, Bologna, Italy.
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy.
- Center for Developmental Biology and Reprogramming-CEDEBIOR, Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy.
| |
Collapse
|
5
|
Park JE, Kim DH. Advanced Immunomodulatory Biomaterials for Therapeutic Applications. Adv Healthc Mater 2024:e2304496. [PMID: 38716543 DOI: 10.1002/adhm.202304496] [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: 12/16/2023] [Revised: 04/15/2024] [Indexed: 05/22/2024]
Abstract
The multifaceted biological defense system modulating complex immune responses against pathogens and foreign materials plays a critical role in tissue homeostasis and disease progression. Recently developed biomaterials that can specifically regulate immune responses, nanoparticles, graphene, and functional hydrogels have contributed to the advancement of tissue engineering as well as disease treatment. The interaction between innate and adaptive immunity, collectively determining immune responses, can be regulated by mechanobiological recognition and adaptation of immune cells to the extracellular microenvironment. Therefore, applying immunomodulation to tissue regeneration and cancer therapy involves manipulating the properties of biomaterials by tailoring their composition in the context of the immune system. This review provides a comprehensive overview of how the physicochemical attributes of biomaterials determine immune responses, focusing on the physical properties that influence innate and adaptive immunity. This review also underscores the critical aspect of biomaterial-based immune engineering for the development of novel therapeutics and emphasizes the importance of understanding the biomaterials-mediated immunological mechanisms and their role in modulating the immune system.
Collapse
Affiliation(s)
- Ji-Eun Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
- Department of Integrative Energy Engineering, College of Engineering, Korea University, Seoul, 02841, Republic of Korea
- Biomedical Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| |
Collapse
|
6
|
Ak A. Fibroblast Cell Responses to Vanadium and Niobium Titanium Alloys: A Biocompatibility Study. ACS OMEGA 2023; 8:33802-33808. [PMID: 37744787 PMCID: PMC10515373 DOI: 10.1021/acsomega.3c04252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023]
Abstract
The interactions of a biomaterial with tissues must be determined for the material to be fully compatible with the body for a long time. The tissue and environment where the material is implanted are highly affected by its content. Titanium-6Aluminum-4Vanadium is widely used in orthopedics and dentistry. Recently, Titanium-6Aluminum-7Niobium alloys have been studied because of Titanium-6Aluminum-4Vanadium toxicity, which may be caused by vanadium. The aim of this study was to determine whether Titanium-6Aluminum-4Vanadium and Titanium-6Aluminum-7Niobium affect fibroblast cell proliferation, mineralization, and collagen production and whether they change the expression of type 1 collagen and fibronectin genes. It was determined that the niobium-containing alloy increased cell proliferation and calcium mineralization compared with the vanadium-containing alloy (p < 0.05). However, the alloys did not cause changes in the expression of collagen type 1 or fibronectin in cells. The collagen content of the cells on the niobium-containing alloy was lower than that on both the vanadium-containing alloy and tissue culture plate surface (p < 0.05). The niobium-containing alloy was found to be superior to the vanadium-containing alloy in terms of cell proliferation and calcium mineralization. Furthermore, neither vanadium-containing alloy nor niobium-containing alloy implant materials altered gene expression. Although both alloys are considered compatible with bone tissue, it should be considered whether they are also biocompatible with fibroblast cells.
Collapse
Affiliation(s)
- Ayse Ak
- Kocaeli Vocational School
of Health Services, Department of Medical Services and Techniques,
Medical Imaging Techniques Program, Kocaeli
University, Kocaeli 41380, Turkey
| |
Collapse
|
7
|
Cremona M, Rusconi G, Ferrario A, Mariotta L, Gola M, Soldati G. Processing Adipose Tissue Samples in a GMP Environment Standardizes the Use of SVF in Cell Therapy Treatments: Data on 302 Patients. Biomedicines 2023; 11:2533. [PMID: 37760974 PMCID: PMC10525825 DOI: 10.3390/biomedicines11092533] [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: 07/11/2023] [Revised: 08/09/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Stromal vascular fraction (SVF) cells, together with adipose-derived mesenchymal stem cells, are becoming the tool of choice for many clinical applications. Currently, nearly 200 clinical trials are running worldwide to prove the efficacy of this cell type in treating many diseases and pathological conditions. To reach the goals of cell therapies and produce ATMPs as drugs for regenerative medicine, it is necessary to properly standardize GMP processes and, thus, collection methods, transportation strategies, extraction protocols, and characterization procedures, without forgetting that all the tissues of the human body are characterized by a wide inter-individual variability which is genetically determined and acquired during life. Here, we compare 302 samples processed under GMP rules to exclude the influence of the operator and of the anatomical site of collection. The influence of variability in the ages and genders of patients, along with laboratory parameters such as total cell number, cell viability, stem cell number, and other stromal vascular fraction cell subpopulations, has been compared. The results show that when the laboratory protocol is standardized, the variability of quantifiable cell parameters is widely statistically non-significant, meaning that we can take a further step toward standardized advanced cell therapy products.
Collapse
Affiliation(s)
- Martina Cremona
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
| | - Giulio Rusconi
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
| | | | - Luca Mariotta
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
- Swiss Stem Cells Biotech AG, 8008 Zürich, Switzerland
| | - Mauro Gola
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
| | - Gianni Soldati
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
| |
Collapse
|
8
|
Obesity and Wound Healing: Focus on Mesenchymal Stem Cells. Life (Basel) 2023; 13:life13030717. [PMID: 36983872 PMCID: PMC10059997 DOI: 10.3390/life13030717] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/20/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
Chronic wounds represent nowadays a major challenge for both clinicians and researchers in the regenerative setting. Obesity represents one of the major comorbidities in patients affected by chronic ulcers and therefore diverse studies aimed at assessing possible links between these two morbid conditions are currently ongoing. In particular, adipose tissue has recently been described as having metabolic and endocrine functions rather than serving as a mere fat storage deposit. In this setting, adipose-derived stem cells, a peculiar subset of mesenchymal stromal/stem cells (MSCs) located in adipose tissue, have been demonstrated to possess regenerative and immunological functions with a key role in regulating both adipocyte function and skin regeneration. The aim of the present review is to give an overview of the most recent findings on wound healing, with a special focus on adipose tissue biology and obesity.
Collapse
|
9
|
Brembilla NC, Vuagnat H, Boehncke WH, Krause KH, Preynat-Seauve O. Adipose-Derived Stromal Cells for Chronic Wounds: Scientific Evidence and Roadmap Toward Clinical Practice. Stem Cells Transl Med 2022; 12:17-25. [PMID: 36571216 PMCID: PMC9887085 DOI: 10.1093/stcltm/szac081] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/16/2022] [Indexed: 12/27/2022] Open
Abstract
Chronic wounds, ie, non-healing ulcers, have a prevalence of ~1% in the general population. Chronic wounds strongly affect the quality of life and generate considerable medical costs. A fraction of chronic wounds will heal within months of appropriate treatment; however, a significant fraction of patients will develop therapy-refractory chronic wounds, leading to chronic pain, infection, and amputation. Given the paucity of therapeutic options for refractory wounds, cell therapy and in particular the use of adipose-derived stromal cells (ASC) has emerged as a promising concept. ASC can be used as autologous or allogeneic cells. They can be delivered in suspension or in 3D cultures within scaffolds. ASC can be used without further processing (stromal vascular fraction of the adipose tissue) or can be expanded in vitro. ASC-derived non-cellular components, such as conditioned media or exosomes, have also been investigated. Many in vitro and preclinical studies in animals have demonstrated the ASC efficacy on wounds. ASC efficiency appears to occurs mainly through their regenerative secretome. Hitherto, the majority of clinical trials focused mainly on safety issues. However more recently, a small number of randomized, well-controlled trials provided first convincing evidences for a clinical efficacy of ASC-based chronic wound therapies in humans. This brief review summarizes the current knowledge on the mechanism of action, delivery and efficacy of ASC in chronic wound therapy. It also discusses the scientific and pharmaceutical challenges to be solved before ASC-based wound therapy enters clinical reality.
Collapse
Affiliation(s)
- Nicolo C Brembilla
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland,Division of Dermatology and Venereology, Geneva University Hospitals, Geneva, Switzerland
| | - Hubert Vuagnat
- Program for Wounds and Wound Healing, Care Directorate, Geneva University Hospitals, Geneva, Switzerland
| | - Wolf-Henning Boehncke
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland,Division of Dermatology and Venereology, Geneva University Hospitals, Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland,Laboratory of Therapy and Stem Cells, Geneva University Hospitals, Geneva, Switzerland
| | - Olivier Preynat-Seauve
- Corresponding author: Olivier Preynat-Seauve, PATIM, 1 rue Michel Servet CH-1211 Geneva 4, Switzerland. Tel: +41223794139;
| |
Collapse
|
10
|
Kanta J, Zavadakova A, Sticova E, Dubsky M. Fibronectin in hyperglycaemia and its potential use in the treatment of diabetic foot ulcers: A review. Int Wound J 2022; 20:1750-1761. [PMID: 36537075 PMCID: PMC10088845 DOI: 10.1111/iwj.13997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 12/24/2022] Open
Abstract
Metabolism of fibronectin, the protein that plays a key role in the healing of wounds, is changed in the patients with diabetes mellitus. Fibronectin can interact with other proteins and proteoglycans and organise them to form the extracellular matrix, the basis of the granulation tissue in healing wounds. However, diabetic foot ulcers (DFUs) suffer from inadequate deposition of this protein. Degradation prevails over fibronectin synthesis in the proteolytic inflammatory environment in the ulcers. Because of the lack of fibronectin in the wound bed, the assembly of the extracellular matrix and the deposition of the granulation tissue cannot be started. A number of methods have been designed that prevents fibronectin degradation, replace lacking fibronectin or support its formation in non-healing wounds in animal models of diabetes. The aim of this article is to review the metabolism of fibronectin in DFUs and to emphasise that it would be useful to pay more attention to fibronectin matrix assembly in the ulcers when laboratory methods are translated to clinical practice.
Collapse
Affiliation(s)
- Jiri Kanta
- Faculty of Medicine Charles University Hradec Kralove Czech Republic
| | - Anna Zavadakova
- Biomedical Center, Faculty of Medicine Charles University Pilsen Czech Republic
| | - Eva Sticova
- Diabetes Center Institute for Clinical and Experimental Medicine Prague Czech Republic
- Third Faculty of Medicine Charles University Prague Czech Republic
| | - Michal Dubsky
- Diabetes Center Institute for Clinical and Experimental Medicine Prague Czech Republic
- First Faculty of Medicine Charles University Prague Czech Republic
| |
Collapse
|
11
|
Adipose Tissue Development Relies on Coordinated Extracellular Matrix Remodeling, Angiogenesis, and Adipogenesis. Biomedicines 2022; 10:biomedicines10092227. [PMID: 36140327 PMCID: PMC9496222 DOI: 10.3390/biomedicines10092227] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/24/2022] Open
Abstract
Despite developing prenatally, the adipose tissue is unique in its ability to undergo drastic growth even after reaching its mature size. This development and subsequent maintenance rely on the proper coordination between the vascular niche and the adipose compartment. In this review, the process of adipose tissue development is broken down to explain (1) the ultrastructural matrix remodeling that is undertaken during simultaneous adipogenesis and angiogenesis, (2) the paracrine crosstalk involved during adipose development, (3) the mechanical regulators involved in adipose growth, and (4) the proteolytic and paracrine oversight for matrix remodeling during adipose development. It is crucial to gain a better understanding of the complex relationships that exist between adipose tissue and the vasculature during tissue development to provide insights into the pathological tissue expansion of obesity and to develop improved soft-tissue reconstruction techniques.
Collapse
|
12
|
Paganelli A, Rossi E, Magnoni C. The dark side of adipose-derived mesenchymal stromal cells in cutaneous oncology: roles, expectations, and potential pitfalls. Stem Cells Dev 2022; 31:593-603. [PMID: 36066334 DOI: 10.1089/scd.2022.0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Adipose-derived stromal cells (ADSCs) have well-established regenerative and immunomodulatory properties. For such reasons, ADSCs are currently under investigation for their use in the setting of both regenerative medicine and autoimmune diseases. As per dermatological disorders, MSC-based strategies represent potential therapeutic tools not only for chronic ulcers and wound healing, but also for immune-mediated dermatoses. However, a growing body of research has been focusing on the role of MSCs in human cancers, due to the potential oncological risk of using MSC-based strategies linked to their anti-apoptotic, pro-angiogenic and immunosuppressive properties. In the dermatological setting, ADSCs have shown not only to promote melanoma growth and invasiveness, but also to induce drug-resistance. On the other hand, genetically modified ADSCs have been demonstrated to efficiently target therapies at tumor sites, due to their migratory properties and their peculiar tropism for cancer microenvironment. The present review briefly summarizes the findings published so far on the use of ADSCs in the dermato-oncological setting, with the majority of data being available for melanoma.
Collapse
Affiliation(s)
- Alessia Paganelli
- Universita degli Studi di Modena e Reggio Emilia, Dermatology, Modena, Italy, 41124;
| | - Elena Rossi
- Universita degli Studi di Modena e Reggio Emilia, Dermatology, Modena, Italy;
| | - Cristina Magnoni
- Universita degli Studi di Modena e Reggio Emilia, Dermatology, Modena, Italy;
| |
Collapse
|
13
|
Zhao J, Bi X, Wu W, Lin Z, Li Y, Chen Y, Lu F, Dong Z. 3D Culture Promotes Secretion of Extracellular Matrix Structure Fat Flap with Lipoaspirates in Vitro. Tissue Eng Part A 2022; 28:929-939. [PMID: 36017621 DOI: 10.1089/ten.tea.2022.0070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Adipose tissue engineering represents a possible solution for large-volume soft-tissue reconstruction. Although there have been several reports on the construction of tissue-engineered fat (TEF) flaps in vivo and in vitro, each condition has various limitations. Thus, we developed a novel approach for engineering fat tissue using a 3D culture system. We used different volumes of lipoaspirates to fill the same tissue engineering chamber (30%, 50%, 80%, and 100% volume/space ratio) for different periods (3, 5, 7, and 14 days) to determine whether lipoaspirates can form structural fat tissue in vitro. We then studied the histological structure and extracellular matrix of the tissue formed in vitro. We selected engineering tissue-like fat of the 80% volume/space ratio group cultured for 7 days to be subcutaneously implanted into mice for up to 3 months, and lipoaspirates without structure in vitro were used as a control. The lipoaspirates from the 80% volume/space ratio group cultured in vitro formed TEF-like tissue, which increased in small adipocytes and extracellular matrix with time until becoming stable on day 7. The live/dead test showed that the tissue cultured in vitro remained viable until day 7. Immunofluorescence staining results revealed that the collagen I and IV content increased over time. Moreover, after grafting, "self-assembly" fat had higher volume retention, better vascularization, fewer oil droplets, and less fibrosis than the lipoaspirates without structure in vitro. Therefore, our results demonstrate that lipoaspirates filled in tissue engineering chamber can be cultured in vitro and can "self-assemble" into TEF-like tissue. Furthermore, the "self-assembly" fat tissue produced better grafting results than those of lipoaspirates without structure in vitro.
Collapse
Affiliation(s)
- Jing Zhao
- Southern Medical University Nanfang Hospital, 1838 Guangzhou North Road, Guangzhou, Guangdong 510515, P.R.China, Guangzhou, Guangdong, China, 510515;
| | - Xin Bi
- Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China;
| | - Weizi Wu
- Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China;
| | - Zhousheng Lin
- Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China;
| | - Ye Li
- Southern Medical University Nanfang Hospital, Plastic and Cosmetic Surgery, guangzhou, Guangzhou, China, 510515;
| | - Yunzi Chen
- Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China;
| | - Feng Lu
- Southern Medical University Nanfang Hospital, Department of Plastic Surgery, Guangzhou, Guangdong, China;
| | - Ziqing Dong
- Southern Medical University Nanfang Hospital, Department of Plastic Surgery, Guangzhou, Guangdong, China;
| |
Collapse
|
14
|
Safoine M, Côté A, Leloup R, Hayward CJ, Plourde Campagna MA, Ruel J, Fradette J. Engineering naturally-derived human connective tissues for clinical applications using a serum-free production system. Biomed Mater 2022; 17. [PMID: 35950736 DOI: 10.1088/1748-605x/ac84b9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 07/27/2022] [Indexed: 11/12/2022]
Abstract
The increasing need for tissue substitutes in reconstructive surgery spurs the development of engineering methods suited for clinical applications. Cell culture and tissue production traditionally require the use of fetal bovine serum (FBS) which is associated with various complications especially from a translational perspective. Using the self-assembly approach of tissue engineering, we hypothesized that all important parameters of tissue reconstruction can be maintained in a production system devoid of FBS from cell extraction to tissue reconstruction. We studied two commercially available serum-free medium (SFM) and xenogen-free serum-free medium (XSFM) for their impact on tissue reconstruction using human adipose-derived stem/stromal cells (ASCs) in comparison to serum-containing medium. Both media allowed higher ASC proliferation rates in primary cultures over five passages compared with 10% FBS supplemented medium while maintaining high expression of mesenchymal cell markers. For both media, we evaluated extracellular matrix production and deposition necessary to engineer manipulatable tissues using the self-assembly approach. Tissues produced in SFM exhibited a significantly increased thickness (up to 6.8-fold) compared with XSFM and FBS-containing medium. A detailed characterization of tissues produced under SFM conditions showed a substantial 50% reduction of production time without compromising key tissue features such as thickness, mechanical resistance and pro-angiogenic secretory capacities (plasminogen activator inhibitor 1, hepatocyte growth factor, vascular endothelial growth factor, angiopoietin-1) when compared to tissues produced in the control FBS-containing medium. Furthermore, we compared ASCs to the frequently used human dermal fibroblasts (DFs) in the SFM culture system. ASC-derived tissues displayed a 2.4-fold increased thickness compared to their DFs counterparts. In summary, we developed all-natural human substitutes using a production system compatible with clinical requirements. Under culture conditions devoid of bovine serum, the resulting engineered tissues displayed similar and even superior structural and functional properties over the classic FBS-containing culture conditions with a considerable 50% shortening of production time.
Collapse
Affiliation(s)
- Meryem Safoine
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada.,Division of Regenerative Medicine, CHU de Québec-Université Laval Research Centre, Québec, QC, Canada
| | - Alexandra Côté
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada.,Division of Regenerative Medicine, CHU de Québec-Université Laval Research Centre, Québec, QC, Canada
| | - Romane Leloup
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada.,Division of Regenerative Medicine, CHU de Québec-Université Laval Research Centre, Québec, QC, Canada
| | - Cindy Jean Hayward
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada.,Division of Regenerative Medicine, CHU de Québec-Université Laval Research Centre, Québec, QC, Canada
| | - Marc-André Plourde Campagna
- Bureau de design, Department of Mechanical Engineering, Faculty of Science and Engineering, Université Laval, Québec, QC, Canada
| | - Jean Ruel
- Division of Regenerative Medicine, CHU de Québec-Université Laval Research Centre, Québec, QC, Canada.,Bureau de design, Department of Mechanical Engineering, Faculty of Science and Engineering, Université Laval, Québec, QC, Canada
| | - Julie Fradette
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada.,Division of Regenerative Medicine, CHU de Québec-Université Laval Research Centre, Québec, QC, Canada.,Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
15
|
Kim S, Lee HY, Lee HR, Jang JY, Yun JH, Shin YS, Kim CH. Liquid-type plasma-controlled in situ crosslinking of silk-alginate injectable gel displayed better bioactivities and mechanical properties. Mater Today Bio 2022; 15:100321. [PMID: 35757030 PMCID: PMC9214807 DOI: 10.1016/j.mtbio.2022.100321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022]
Abstract
Silk is a promising biomaterial for injectable hydrogel, but its long-gelation time and cytotoxic crosslinking methods are the main obstacles for clinical application. Here, we purpose a new in situ crosslinking technique of silk-alginate (S-A) injectable hydrogel using liquid-type non-thermal atmospheric plasma (LTP) in vocal fold (VF) wound healing. We confirmed that LTP induces the secondary structure of silk in a dose-dependent manner, resulting in improved mechanical properties. Significantly increased crosslinking of silk was observed with reduced gelation time. Moreover, controlled release of nitrate, an LTP effectors, from LTP-treated S-A hydrogel was detected over 7 days. In vitro experiments regarding biocompatibility showed activation of fibroblasts beyond the non-cytotoxicity of LTP-treated S-A hydrogels. An in vivo animal model of VF injury was established in New Zealand White rabbits. Full-thickness injury was created on the VF followed by hydrogel injection. In histologic analyses, LTP-treated S-A hydrogels significantly reduced a scar formation and promoted favorable wound healing. Functional analysis using videokymography showed eventual viscoelastic recovery. The LTP not only changes the mechanical structures of a hydrogel, but also has sustained biochemical effects on the damaged tissue due to controlled release of LTP effectors, and that LTP-treated S-A hydrogel can be used to enhance wound healing after VF injury.
Collapse
Affiliation(s)
- Sungryeal Kim
- Department of Otolaryngology, College of Medicine, Inha University, Incheon, South Korea.,Department of Medical Sciences, Graduate School of Ajou University, Suwon, South Korea
| | - Hye-Young Lee
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, South Korea
| | - Hye Ran Lee
- Department of Otorhino-laryngology-Head and Neck Surgery, Catholic Kwandong University, College of Medicine, Incheon, South Korea
| | - Jeon Yeob Jang
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, South Korea
| | - Ju Hyun Yun
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, South Korea
| | - Yoo Seob Shin
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, South Korea
| | - Chul-Ho Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, South Korea
| |
Collapse
|
16
|
Paganelli A, Benassi L, Rossi E, Tarentini E, Magnoni C. Mesenchymal stromal cells promote the proliferation of basal stem cells and efficient epithelization in organotypic models of wound healing. Microsc Res Tech 2022; 85:2752-2756. [PMID: 35388560 PMCID: PMC9322434 DOI: 10.1002/jemt.24110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 03/05/2022] [Accepted: 03/20/2022] [Indexed: 12/21/2022]
Abstract
Adipose derived mesenchymal stromal cells (ADSCs) represent a fascinating tool in the scenario of wound healing and regenerative medicine. Recent data already demonstrated that ADSCs could exert a stimulatory action on epithelial cells through secretion of soluble factors. The aim of the present study was to assess how ADSCs guide wound re‐epithelization in vitro in the presence of keratinocytes. We used an organotypic model of wound healing and we seeded keratinocytes on a ADSC‐induced dermal matrix. Conventional hematoxylin–eosin stain and immunohistochemistry staining for Ki67, p63 and pan‐keratins were performed at different timepoints. Histological sections of organotypic cultures showed complete coverage of the ADSC‐induced matrix by keratinocytes. Proliferation of basal stem cells was found to be the main mechanism responsible for epithelization of the dermis. In conclusion, ADSC do not only stimulate dermal regeneration through collagen deposition but also promote epithelization.
Collapse
Affiliation(s)
- Alessia Paganelli
- Surgical, Medical and Dental Department of Morphological Sciences related to Transplant, Oncology and Regenerative Medicine, Division of Dermatology, University of Modena and Reggio Emilia, Modena, Italy.,PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Luisa Benassi
- Surgical, Medical and Dental Department of Morphological Sciences related to Transplant, Oncology and Regenerative Medicine, Division of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Rossi
- Surgical, Medical and Dental Department of Morphological Sciences related to Transplant, Oncology and Regenerative Medicine, Division of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Tarentini
- Surgical, Medical and Dental Department of Morphological Sciences related to Transplant, Oncology and Regenerative Medicine, Division of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Magnoni
- Surgical, Medical and Dental Department of Morphological Sciences related to Transplant, Oncology and Regenerative Medicine, Division of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
17
|
Bioinks Enriched with ECM Components Obtained by Supercritical Extraction. Biomolecules 2022; 12:biom12030394. [PMID: 35327586 PMCID: PMC8945720 DOI: 10.3390/biom12030394] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 02/01/2023] Open
Abstract
Extracellular matrix (ECM)-based bioinks have been steadily gaining interest in the field of bioprinting to develop biologically relevant and functional tissue constructs. Herein, we propose the use of supercritical carbon dioxide (scCO2) technology to extract the ECM components of cell-sheets that have shown promising results in creating accurate 3D microenvironments replicating the cell’s own ECM, to be used in the preparation of bioinks. The ECM extraction protocol best fitted for cell sheets was defined by considering efficient DNA removal with a minor effect on the ECM. Cell sheets of human dermal fibroblasts (hDFbs) and adipose stem cells (hASCs) were processed using a customised supercritical system by varying the pressure of the reactor, presence, exposure time, and type of co-solvent. A quantification of the amount of DNA, protein, and sulfated glycosaminoglycans (sGAGs) was carried out to determine the efficiency of the extraction in relation to standard decellularization methodologies. The bioinks containing the extracted ECM were fabricated by combining them with alginate as a support polymer. The influence of the alginate (1%, 2% w/vol) and ECM (0.5% and 1.5% w/vol) amounts on the printability of the blends was addressed by analysing the rheological behaviour of the suspensions. Finally, 3D printed constructs were fabricated using an in-house built extrusion-based bioprinter, and the impact of the extrusion process on cell viability was assessed. The optimised scCO2 protocol allowed efficient removal of DNA while preserving a higher number of proteins and sGAGs than the standard methodologies. The characterization of extract’s composition also revealed that the ECM produced by hDFbs (fECM) and hASCs (aECM) is distinctively affected by the extraction protocols. Furthermore, rheological analysis indicated an increase in viscosity with increasing ECM composition, an effect even more prominent in samples containing aECM. 3D printing of alginate/ECM constructs demonstrated that cell viability was only marginally affected by the extrusion process, and this effect was also dependent on the ECM source. Overall, this work highlights the benefits of supercritical fluid-based methods for ECM extraction and strengthens the relevance of ECM-derived bioinks in the development of printed tissue-like constructs.
Collapse
|
18
|
Guan Y, Yang B, Xu W, Li D, Wang S, Ren Z, Zhang J, Zhang T, Liu XZ, Li J, Li C, Meng F, Han F, Wu T, Wang Y, Peng J. Cell-derived extracellular matrix materials for tissue engineering. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:1007-1021. [PMID: 34641714 DOI: 10.1089/ten.teb.2021.0147] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The involvement of cell-derived extracellular matrix (CDM) in assembling tissue engineering scaffolds has yielded significant results. CDM possesses excellent characteristics, such as ideal cellular microenvironment mimicry and good biocompatibility, which make it a popular research direction in the field of bionanomaterials. CDM has significant advantages as an expansion culture substrate for stem cells, including stabilization of phenotype, reversal of senescence, and guidance of specific differentiation. In addition, the applications of CDM-assembled tissue engineering scaffolds for disease simulation and tissue organ repair are comprehensively summarized; the focus is mainly on bone and cartilage repair, skin defect or wound healing, engineered blood vessels, peripheral nerves, and periodontal tissue repair. We consider CDM a highly promising bionic biomaterial for tissue engineering applications and propose a vision for its comprehensive development.
Collapse
Affiliation(s)
- Yanjun Guan
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Boyao Yang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Wenjing Xu
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Dongdong Li
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Sidong Wang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Zhiqi Ren
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Jian Zhang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Tieyuan Zhang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Xiu-Zhi Liu
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Junyang Li
- Nankai University School of Medicine, 481107, Tianjin, Tianjin, China.,Chinese PLA General Hospital, 104607, Beijing, Beijing, China;
| | - Chaochao Li
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Fanqi Meng
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China.,Peking University People's Hospital, 71185, Department of spine surgery, Beijing, China;
| | - Feng Han
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Tong Wu
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Yu Wang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China.,Nantong University, 66479, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China;
| | - Jiang Peng
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China.,Nantong University, 66479, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China;
| |
Collapse
|
19
|
Paganelli A, Tarentini E, Benassi L, Scelfo D, Pisciotta A, Rossi E, Magnoni C. Use of confocal microscopy imaging for in vitro assessment of adipose-derived mesenchymal stromal cells seeding on acellular dermal matrices: 3D reconstruction based on collagen autofluorescence. Skin Res Technol 2021; 28:133-141. [PMID: 34555218 PMCID: PMC9292443 DOI: 10.1111/srt.13103] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/21/2021] [Indexed: 12/16/2022]
Abstract
Background Both mesenchymal stromal cells (MSCs) and acellular dermal matrices (ADMs) represent fascinating therapeutic tools in the wound healing scenario. Strategies aimed at combining these two treatment modalities are currently under investigation. Moreover, scarcity of quantitative, nondestructive techniques for quality assessment of engineered tissues poses great limitations in regenerative medicine and collagen autofluorescence‐based imaging techniques are acquiring great importance in this setting. Objective Our goals were to assess the in vitro interactions between ADSCs and ADMs and to analyze extracellular‐matrix production. Methods Adipose‐derived MSCs (ADSC) were plated on 8‐mm punch biopsies of a commercially available ADM (Integra®). Conventional histology with hematoxylin‐eosin staining, environmental scanning electron microscopy, and confocal‐laser scanning microscopy were used to obtain imaging of ADSC‐seeded ADMs. Collagen production by ADSCs was quantified by mean fluorescence intensity (MFI), expressed in terms of positive pixels/field, obtained through ImageJ software processing of three‐dimensional projections from confocal scanning images. Control conditions included: fibroblast‐seeded ADM, ADSC‐ and fibroblast‐induced scaffolds, and Integra® alone. Results ADSCs were efficiently seeded on Integra® and were perfectly incorporated in the pores of the scaffold. Collagen production was revealed to be significantly higher when ADSCs were seeded on ADM rather than in all other control conditions. Collagen autofluorescence was efficiently used as a surrogate marker of ECM production. Conclusions Combined therapies based on MSCs and collagenic ADMs are promising therapeutic options for chronic wounds. Not only ADSCs can be efficiently seeded on ADMs, but ADMs also seem to potentiate their regenerative properties, as highlightable from fluorescence confocal imaging.
Collapse
Affiliation(s)
- Alessia Paganelli
- Surgical, Medical and Dental Department of Morphological Sciences related to Transplant, Oncology and Regenerative Medicine, Division of Dermatology, University of Modena and Reggio Emilia, Modena and Reggio Emilia, Italy.,PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena and Reggio Emilia, Italy
| | - Elisabetta Tarentini
- Surgical, Medical and Dental Department of Morphological Sciences related to Transplant, Oncology and Regenerative Medicine, Division of Dermatology, University of Modena and Reggio Emilia, Modena and Reggio Emilia, Italy
| | - Luisa Benassi
- Surgical, Medical and Dental Department of Morphological Sciences related to Transplant, Oncology and Regenerative Medicine, Division of Dermatology, University of Modena and Reggio Emilia, Modena and Reggio Emilia, Italy
| | - Daniel Scelfo
- Surgical, Medical and Dental Department of Morphological Sciences related to Transplant, Oncology and Regenerative Medicine, Division of Dermatology, University of Modena and Reggio Emilia, Modena and Reggio Emilia, Italy
| | - Alessandra Pisciotta
- Surgical, Medical and Dental Department of Morphological Sciences related to Transplant, Oncology and Regenerative Medicine, Division of Dermatology, University of Modena and Reggio Emilia, Modena and Reggio Emilia, Italy
| | - Elena Rossi
- Surgical, Medical and Dental Department of Morphological Sciences related to Transplant, Oncology and Regenerative Medicine, Division of Dermatology, University of Modena and Reggio Emilia, Modena and Reggio Emilia, Italy
| | - Cristina Magnoni
- Surgical, Medical and Dental Department of Morphological Sciences related to Transplant, Oncology and Regenerative Medicine, Division of Dermatology, University of Modena and Reggio Emilia, Modena and Reggio Emilia, Italy
| |
Collapse
|
20
|
Lee HR, Lee HY, Heo J, Jang JY, Shin YS, Kim CH. Liquid-type nonthermal atmospheric plasma enhanced regenerative potential of silk-fibrin composite gel in radiation-induced wound failure. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112304. [PMID: 34474855 DOI: 10.1016/j.msec.2021.112304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 06/29/2021] [Accepted: 07/04/2021] [Indexed: 12/15/2022]
Abstract
Delayed wound healing in heavily irradiated areas is a serious clinical complication that makes widespread therapeutic use of radiation difficult. Efficient treatment strategies are urgently required for addressing radiation-induced wound failure. Herein, we applied liquid-type nonthermal atmospheric plasma (LTP) to a silk-fibrin (SF) composite gel to investigate whether controlled release of LTP from SF hydrogel not only induced favorable cellular events in an irradiated wound bed but also modulated the SF hydrogel microstructure itself, eventually facilitating the development of a regenerative microenvironment. Scanning electron microscopy and Fourier-transform infrared spectroscopy revealed that LTP modulated the microstructures and chemical bindings of the SF gel. Improved cell viability, morphology, and extracellular matrix depositions by the LTP-treated SF hydrogel were identified with wound-healing assays and immunofluorescence staining. An irradiated random-pattern skin-flap animal model was established in six-week-old C57/BL6 mice. Full-thickness skin was flapped from the dorsum and SF hydrogel was placed underneath the raised skin flap. Postoperative histological analysis of the irradiated random-pattern skin-flap mice model suggested that LTP-treated SF hydrogel much improved wound regeneration and the inflammatory response compared to the SF hydrogel- and sham-treated groups. These results support that LTP-treated SF hydrogel significantly enhanced irradiated wound healing. Cellular and tissue reactions to released LTP from the SF hydrogel were favorable for the regenerative process of the wound; furthermore, mechanochemical properties of the SF gel were improved by LTP.
Collapse
Affiliation(s)
- Hye Ran Lee
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | - Hye-Young Lee
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | - Jaesung Heo
- Department of Radiation Oncology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | - Jeon Yeob Jang
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | - Yoo Seob Shin
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea.
| | - Chul-Ho Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea
| |
Collapse
|
21
|
François P, Rusconi G, Arnaud L, Mariotta L, Giraudo L, Minonzio G, Veran J, Bertrand B, Dumoulin C, Grimaud F, Lyonnet L, Casanova D, Giverne C, Cras A, Magalon G, Dignat-George F, Sabatier F, Magalon J, Soldati G. Inter-center comparison of good manufacturing practices-compliant stromal vascular fraction and proposal for release acceptance criteria: a review of 364 productions. Stem Cell Res Ther 2021; 12:373. [PMID: 34210363 PMCID: PMC8252207 DOI: 10.1186/s13287-021-02445-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/10/2021] [Indexed: 11/10/2022] Open
Abstract
Background Even though the manufacturing processes of the stromal vascular fraction for clinical use are performed in compliance with the good manufacturing practices applying to advanced therapy medicinal products, specifications related to stromal vascular fraction quality remain poorly defined. We analyzed stromal vascular fraction clinical batches from two independent good manufacturing practices-compliant manufacturing facilities, the Swiss Stem Cell Foundation (SSCF) and Marseille University Hospitals (AP-HM), with the goal of defining appropriate and harmonized release acceptance criteria. Methods This retrospective analysis reviewed the biological characteristics of 364 batches of clinical-grade stromal vascular fraction. Collected data included cell viability, recovery yield, cell subset distribution of stromal vascular fraction, and microbiological quality. Results Stromal vascular fraction from SSCF cohort demonstrated a higher viability (89.33% ± 4.30%) and recovery yield (2.54 × 105 ± 1.22 × 105 viable nucleated cells (VNCs) per mL of adipose tissue) than stromal vascular fraction from AP-HM (84.20% ± 5.96% and 2.25 × 105 ± 1.11 × 105 VNCs per mL). AP-HM batches were significantly less contaminated (95.71% of sterile batches versus 74.15% for SSCF batches). The cell subset distribution was significantly different (higher proportion of endothelial cells and lower proportion of leukocytes and pericytes in SSCF cohort). Conclusions Both centers agreed that a good manufacturing practices-compliant stromal vascular fraction batch should exert a viability equal or superior to 80%, a minimum recovery yield of 1.50 × 105 VNCs per mL of adipose tissue, a proportion of adipose-derived stromal cells at least equal to 20%, and a proportion of leukocytes under 50%. In addition, a multiparameter gating strategy for stromal vascular fraction analysis is proposed. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02445-z.
Collapse
Affiliation(s)
- Pauline François
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 147 Bd Baille, 13005, Marseille, France.,Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France
| | - Giulio Rusconi
- Swiss Stem Cell Foundation, Gentilino, Lugano, Switzerland.,Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Laurent Arnaud
- Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Luca Mariotta
- Swiss Stem Cell Foundation, Gentilino, Lugano, Switzerland
| | - Laurent Giraudo
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 147 Bd Baille, 13005, Marseille, France
| | - Greta Minonzio
- Swiss Stem Cell Foundation, Gentilino, Lugano, Switzerland
| | - Julie Veran
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 147 Bd Baille, 13005, Marseille, France
| | - Baptiste Bertrand
- Plastic Surgery Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Chloé Dumoulin
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 147 Bd Baille, 13005, Marseille, France
| | - Fanny Grimaud
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 147 Bd Baille, 13005, Marseille, France
| | - Luc Lyonnet
- Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Dominique Casanova
- Plastic Surgery Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Camille Giverne
- Normandie Univ, UNIROUEN, INSERM, U1234, Rouen University Hospital, Department of Immunology and Biotherapy, Rouen, France
| | - Audrey Cras
- Assistance Publique-Hôpitaux de Paris, Saint-Louis Hospital, Cell Therapy Unit, Cord blood Bank and CIC-BT501, Paris, France
| | | | - Françoise Dignat-George
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France.,Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Florence Sabatier
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 147 Bd Baille, 13005, Marseille, France.,Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France.,Remedex, Marseille, France
| | - Jeremy Magalon
- Cell Therapy Department, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, 147 Bd Baille, 13005, Marseille, France. .,Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France. .,Remedex, Marseille, France.
| | - Gianni Soldati
- Swiss Stem Cell Foundation, Gentilino, Lugano, Switzerland
| |
Collapse
|
22
|
Li N, Bai B, Zhang H, Zhang W, Tang S. Adipose stem cell secretion combined with biomaterials facilitates large-area wound healing. Regen Med 2020; 15:2311-2323. [PMID: 33320721 DOI: 10.2217/rme-2020-0086] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Adipose-derived stem cell (ADSC)-based therapeutic strategies are in fast-pace advancement in wound treatment due to their availability and the ability to self-renew, undergo multilineage differentiation and self-renewal. Existing studies have successfully explored ADSCs to facilitate scar-free healing of small wounds, but whether the healing of large-area wounds that exhibit over 50% of skin tissue loss in the entire body could be achieved remains controversial. This study sought to review the mechanism of physiological wound healing, and discuss the roles played by chemokines, biological factors and biomaterial scaffolds. The possibility of applying ADSC-conditioned medium or ADSC-released exosomes as 'off-the-shelf' tissue engineering products, integrated with biomaterial scaffolds to facilitate wound healing, was analyzed.
Collapse
Affiliation(s)
- Nan Li
- Institute of Plastic Surgery, Weifang Medical University, No. 4948, Shenglidong Street, Kuiwen District of Weifang City, Shandong Province, PR China
| | - Baoshuai Bai
- Institute of Plastic Surgery, Weifang Medical University, No. 4948, Shenglidong Street, Kuiwen District of Weifang City, Shandong Province, PR China
| | - Hairong Zhang
- Institute of Plastic Surgery, Weifang Medical University, No. 4948, Shenglidong Street, Kuiwen District of Weifang City, Shandong Province, PR China
| | - Wei Zhang
- Institute of Plastic Surgery, Weifang Medical University, No. 4948, Shenglidong Street, Kuiwen District of Weifang City, Shandong Province, PR China
| | - Shengjian Tang
- Institute of Plastic Surgery, Weifang Medical University, No. 4948, Shenglidong Street, Kuiwen District of Weifang City, Shandong Province, PR China
| |
Collapse
|
23
|
De Pieri A, Rana S, Korntner S, Zeugolis DI. Seaweed polysaccharides as macromolecular crowding agents. Int J Biol Macromol 2020; 164:434-446. [PMID: 32679331 DOI: 10.1016/j.ijbiomac.2020.07.087] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/30/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023]
Abstract
Development of mesenchymal stem cell-based tissue engineered implantable devices requires prolonged in vitro culture for the development of a three-dimensional implantable device, which leads to phenotypic drift, thus hindering the clinical translation and commercialisation of such approaches. Macromolecular crowding, a biophysical phenomenon based on the principles of excluded-volume effect, dramatically accelerates and increases extracellular matrix deposition during in vitro culture. However, the optimal macromolecular crowder is still elusive. Herein, we evaluated the biophysical properties of various concentrations of different seaweed in origin sulphated polysaccharides and their effect on human adipose derived stem cell cultures. Carrageenan, possibly due to its high sulphation degree, exhibited the highest negative charge values. No correlation was observed between the different concentrations of the crowders and charge, polydispersity index, hydrodynamic radius and fraction volume occupancy across all crowders. None of the crowders, but arabinogalactan, negatively affected cell viability. Carrageenan, fucoidan, galactofucan and ulvan increased extracellular matrix (especially collagen type I and collagen type V) deposition. Carrageenan induced the highest osteogenic effect and galactofucan and fucoidan demonstrated the highest chondrogenic effect. All crowders were relatively ineffective with respect to adipogenesis. Our data highlight the potential of sulphated seaweed polysaccharides for tissue engineering purposes.
Collapse
Affiliation(s)
- Andrea De Pieri
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Proxy Biomedical Ltd., Coilleach, Spiddal, Galway, Ireland
| | - Shubhasmin Rana
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Stefanie Korntner
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland.
| |
Collapse
|
24
|
Al-Hassan JM, Hinek A, Renno WM, Wang Y, Liu YF, Guan R, Wen XY, Litvack ML, Lindenmaier A, Afzal M, Paul B, Oommen S, Nair D, Kumar J, Khan MA, Palaniyar N, Pace-Asciak C. Potential Mechanism of Dermal Wound Treatment With Preparations From the Skin Gel of Arabian Gulf Catfish: A Unique Furan Fatty Acid (F6) and Cholesta-3,5-Diene (S5) Recruit Neutrophils and Fibroblasts to Promote Wound Healing. Front Pharmacol 2020; 11:899. [PMID: 32625093 PMCID: PMC7314935 DOI: 10.3389/fphar.2020.00899] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/02/2020] [Indexed: 12/19/2022] Open
Abstract
Preparations from Arabian Gulf catfish (Arius bilineatus, Val) epidermal gel secretion (PCEGS) effectively heal chronic wounds in diabetic patients. However, specific lipid components of PCEGS that are responsible for various aspects of wound healing are unknown. Here, we report for the first time that, i) a unique preparation containing only proteins and lipids (Fraction B, FB), derived from the PCEGS accelerated the healing of experimental dermal wounds in female rats (transdermal punch biopsy) in vivo. Histological analyses showed that topical treatment of these wounds with FB promoted the migration of fibroblasts, facilitated the production of extracellular matrix (collagen, fibronectin), induced capillary formation and recruitment of immune cells, and accelerated overall wound healing by day 4 (tested at 1, 2, 3, 4, and 10 days; n=15 for vehicle; n=15 for FB treatment), ii) the lipids responsible for different stages of wound healing were separated into a protein-free bioactive lipid fraction, Ft, which contained a few common long-chain fatty acids, a unique furan fatty acid (F6) and a cholesterol metabolite, cholesta-3,5-diene (S5). Ft (the partially purified lipid fraction of PCEGS), and F6 and S5 present in Ft, proved to be bioactive for wound healing in human dermal fibroblasts. Ft increased the production and extracellular deposition of collagen and fibronectin, ex vivo, iii) Ft and its subcomponents, pure F6 and S5, also promoted human dermal fibroblast migration into the scratch wound gaps, ex vivo, iv) Ft, F6, and S5 promoted the recruitment of neutrophils (Green fluorescence protein labeled) to the site of injury in the transected tailfins of transgenic zebrafish, in vivo, v) Ft, but not F6 or S5, promoted the regeneration of tissues at the wound site in the transgenic zebrafish tailfin, in vivo. Therefore, we conclude that lipid fraction Ft from PCEGS contains the components necessary to promote complete wound healing, and F6 and S5 are responsible for promoting fibroblast and neutrophil recruitment to the site of wounds.
Collapse
Affiliation(s)
- Jassim M Al-Hassan
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| | - Aleksander Hinek
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning (PGCRL), The Hospital for Sick Children, Toronto, ON, Canada
| | - Waleed M Renno
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Yanting Wang
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning (PGCRL), The Hospital for Sick Children, Toronto, ON, Canada
| | - Yuan Fang Liu
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning (PGCRL), The Hospital for Sick Children, Toronto, ON, Canada
| | - Rui Guan
- Departments of Lab Medicine and Pathobiology, and Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Xiao-Yen Wen
- Zebrafish Centre for Advanced Drug Discovery & Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada.,Departments of Lab Medicine and Pathobiology, and Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Michael L Litvack
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning (PGCRL), The Hospital for Sick Children, Toronto, ON, Canada
| | - Andras Lindenmaier
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning (PGCRL), The Hospital for Sick Children, Toronto, ON, Canada
| | - Mohammad Afzal
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| | - Bincy Paul
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| | | | - Divya Nair
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| | - Jijin Kumar
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Meraj A Khan
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning (PGCRL), The Hospital for Sick Children, Toronto, ON, Canada
| | - Nades Palaniyar
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning (PGCRL), The Hospital for Sick Children, Toronto, ON, Canada.,Departments of Lab Medicine and Pathobiology, and Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Cecil Pace-Asciak
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning (PGCRL), The Hospital for Sick Children, Toronto, ON, Canada.,Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
25
|
Theodoro V, de Oliveira Fujii L, Lucke LD, Bortolazzo FO, Silva DFD, Carneiro GD, do Amaral MEC, de Oliveira CA, de Andrade TAM, Bombeiro AL, Vicente CP, do Bomfim FRC, de Oliveira ALR, Bagnato VS, Esquisatto MAM, Mendonça FAS, Dos Santos GMT, de Aro AA. Inhibitory effect of red LED irradiation on fibroblasts and co-culture of adipose-derived mesenchymal stem cells. Heliyon 2020; 6:e03882. [PMID: 32426535 PMCID: PMC7226671 DOI: 10.1016/j.heliyon.2020.e03882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/27/2020] [Accepted: 04/27/2020] [Indexed: 12/29/2022] Open
Abstract
The objective of this study was to evaluate the effects of red Light Emiting Diode (red LED) irradiation on fibroblasts in adipose-derived mesenchymal stem cells (ASC) co-culture on the scratch assay. We hypothesized that red LED irradiation could stimulate paracrine secretion of ASC, contributing to the activation of genes and molecules involved in cell migration and tissue repair. ASC were co-cultured with NIH/3T3 fibroblasts through direct contact and subjected to red LED irradiation (1.45 J/cm2/5min6s) after the scratch assay, during 4 days. Four groups were established: fibroblasts (F), fibroblasts + LED (FL), fibroblasts + ASC (FC) and fibroblasts + LED + ASC (FLC). The analyzes were based on Ctgf and Reck expression, quantification of collagen types I and III, tenomodulin, VEGF, TGF-β1, MMP-2 and MMP-9, as well as viability analysis and cell migration. Higher Ctgf expression was observed in FC compared to F. Group FC presented higher amount of tenomodulin and VEGF in relation to the other groups. In the cell migration analysis, a higher number of cells was observed in the scratched area of the FC group on the 4th day. There were no differences between groups considering cell viability, Reck expression, amount of collagen types I and III, MMP-2 and TGF-β1, whereas TGF-β1 was not detected in the FC group and the MMP-9 in none of the groups. Our hypothesis was not supported by the results because the red LED irradiation decreased the healing response of ASC. An inhibitory effect of the LED irradiation associated with ASC co-culture was observed with reduction of the amount of TGF-β1, VEGF and tenomodulin, possibly involved in the reduced cell migration. In turn, the ASC alone seem to have modulated fibroblast behavior by increasing Ctgf, VEGF and tenomodulin, leading to greater cell migration. In conclusion, red LED and ASC therapy can have independent effects on fibroblast wound healing, but the combination of both does not have a synergistic effect. Therefore, future studies with other parameters of red LED associated with ASC should be tested aiming clinical application for tissue repair.
Collapse
Affiliation(s)
- Viviane Theodoro
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation / FHO, Araras, São Paulo, Brazil
| | - Lucas de Oliveira Fujii
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation / FHO, Araras, São Paulo, Brazil
| | - Leticia Dudri Lucke
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, Campinas, São Paulo, Brazil
| | - Fernanda Oriani Bortolazzo
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, Campinas, São Paulo, Brazil
| | | | - Giane Daniela Carneiro
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, Campinas, São Paulo, Brazil
| | | | - Camila Andréa de Oliveira
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation / FHO, Araras, São Paulo, Brazil
| | | | - André Luis Bombeiro
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, Campinas, São Paulo, Brazil
| | - Cristina Pontes Vicente
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, Campinas, São Paulo, Brazil
| | | | | | | | | | | | - Gláucia Maria Tech Dos Santos
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation / FHO, Araras, São Paulo, Brazil
| | - Andrea Aparecida de Aro
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation / FHO, Araras, São Paulo, Brazil.,Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, Campinas, São Paulo, Brazil
| |
Collapse
|