1
|
de Sousa Moreira A, Lopes B, Sousa AC, Coelho A, Sousa P, Araújo A, Delgado E, Alvites R, Maurício AC. Stem Cell-Based Therapies for Glaucoma Treatment: A Review Bridging the Gap in Veterinary Patients. Int J Mol Sci 2024; 26:232. [PMID: 39796087 PMCID: PMC11719664 DOI: 10.3390/ijms26010232] [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: 11/12/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Retinal diseases are characterized by progressive damage to retinal cells, leading to irreversible vision loss. Among these, glaucoma stands out as a multifactorial neurodegenerative disease involving elevated intraocular pressure, retinal ganglion cell apoptosis, and optic nerve damage, ultimately resulting in blindness in both humans and dogs. Stem cell-based therapies have emerged as a promising therapeutic option for such conditions due to their regenerative and neuroprotective potential. These therapies, particularly those based on mesenchymal stem cells, offer the potential to repair and protect retinal tissues through the bioactive molecules (growth factors, cytokines, chemokines) secreted, their secretome. However, research in this field, especially on the use of umbilical cord mesenchymal stem cells' secretome, remains sparse. Most clinical trials focus on human glaucomatous patients, leaving a significant gap in veterinary patients' application, especially in dogs, with additional research being needed to determine its usefulness in canine glaucoma treatment. Future studies should aim to evaluate these therapies across both human and veterinary contexts, broadening treatment possibilities for glaucoma.
Collapse
Affiliation(s)
- Alícia de Sousa Moreira
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Bruna Lopes
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Ana Catarina Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - André Coelho
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Patrícia Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Ana Araújo
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Esmeralda Delgado
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
- Centro de Investigação Interdisciplinar em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Rui Alvites
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
- Instituto Universitário de Ciências da Saúde (CESPU), Avenida Central de Gandra n° 1317, 4585-116 Paredes, Portugal
| | - Ana Colette Maurício
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| |
Collapse
|
2
|
Mansor NI, Balqis TN, Lani MN, Lye KL, Nor Muhammad NA, Ismail WIW, Abidin SZ. Nature's Secret Neuro-Regeneration Pathway in Axolotls, Polychaetes and Planarians for Human Therapeutic Target Pathways. Int J Mol Sci 2024; 25:11904. [PMID: 39595973 PMCID: PMC11593954 DOI: 10.3390/ijms252211904] [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: 10/03/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Despite significant improvements in the comprehension of neuro-regeneration, restoring nerve injury in humans continues to pose a substantial therapeutic difficulty. In the peripheral nervous system (PNS), the nerve regeneration process after injury relies on Schwann cells. These cells play a crucial role in regulating and releasing different extracellular matrix proteins, including laminin and fibronectin, which are essential for facilitating nerve regeneration. However, during regeneration, the nerve is required to regenerate for a long distance and, subsequently, loses its capacity to facilitate regeneration during this progression. Meanwhile, it has been noted that nerve regeneration has limited capabilities in the central nervous system (CNS) compared to in the PNS. The CNS contains factors that impede the regeneration of axons following injury to the axons. The presence of glial scar formation results from this unfavourable condition, where glial cells accumulate at the injury site, generating a physical and chemical barrier that hinders the regeneration of neurons. In contrast to humans, several species, such as axolotls, polychaetes, and planarians, possess the ability to regenerate their neural systems following amputation. This ability is based on the vast amount of pluripotent stem cells that have the remarkable capacity to differentiate and develop into any cell within their body. Although humans also possess these cells, their numbers are extremely limited. Examining the molecular pathways exhibited by these organisms has the potential to offer a foundational understanding of the human regeneration process. This review provides a concise overview of the molecular pathways involved in axolotl, polychaete, and planarian neuro-regeneration. It has the potential to offer a new perspective on therapeutic approaches for neuro-regeneration in humans.
Collapse
Affiliation(s)
- Nur Izzati Mansor
- Department of Nursing, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia;
| | - Tengku Nabilatul Balqis
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia; (T.N.B.); (W.I.W.I.)
| | - Mohd Nizam Lani
- Faculty of Fisheries and Food Science, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia;
| | - Kwan Liang Lye
- ME Scientifique Sdn Bhd, Taman Universiti Indah, Seri Kembangan 43300, Selangor, Malaysia;
| | - Nor Azlan Nor Muhammad
- Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia;
| | - Wan Iryani Wan Ismail
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia; (T.N.B.); (W.I.W.I.)
- Research Interest Group Biological Security and Sustainability (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia
| | - Shahidee Zainal Abidin
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia; (T.N.B.); (W.I.W.I.)
- Research Interest Group Biological Security and Sustainability (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia
| |
Collapse
|
3
|
Kiuchi S, Lopes TJ, Oishi T, Cho Y, Ochiai H, Gomi T. TSG-6 Is Involved in Fibrous Structural Remodeling after the Injection of Adipose-derived Stem Cells. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2024; 12:e5990. [PMID: 39036595 PMCID: PMC11259393 DOI: 10.1097/gox.0000000000005990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/29/2024] [Indexed: 07/23/2024]
Abstract
Background Although aesthetic treatments can rejuvenate the skin, they often cause specific forms of tissue damage. Unlike wounding, which typically results in fibrotic scar tissue, damage from aesthetic treatments induces a distinct histological rejuvenation. The mechanisms that drive this rejuvenation are not yet fully understood. Here, we were interested in cellular responses following aesthetic treatments injecting adipose-derived stem cells (ASCs) subcutaneously. Through investigation with an ex vivo experimental model, a key gene was identified that orchestrates fibrous structural changes and tissue remodeling. Methods Using fresh human subcutaneous adipose tissue co-cultured with ASCs, the changes in the fibrous architecture of the tissue were sequentially mapped. The key regulatory genes involved in remodeling were identified using gene expression and computational analyses. Results We identified the regulatory elements that are crucial for tissue remodeling. Among those, we found that tumor necrosis factor-stimulated gene-6 (TSG-6) is a paracrine mediator essential for the collagen activity. It not only alleviates tissue inflammation but also promotes collagen replacement ex vivo. This is primarily achieved by inhibiting the formation of neutrophil extracellular traps, which are known to promote fibrosis. Conclusions TSG-6 is a key factor modulating tissue inflammation. As our results demonstrate, after ASCs treatment, this factor directs skin healing away from fibrosis by reducing neutrophil extracellular trap formation in subcutaneous adipose tissue and promotes fibrous rejuvenation.
Collapse
Affiliation(s)
- Satomi Kiuchi
- From POLA Chemical Industries, Inc., Yokohama, Japan
| | - Tiago J.S. Lopes
- Center of Regenerative Medicine, National Center for Child Health and Development Research Institute, Tokyo, Japan
- Nezu Life Sciences, Karlsruhe, Germany
| | - Takaya Oishi
- From POLA Chemical Industries, Inc., Yokohama, Japan
| | - Yuki Cho
- From POLA Chemical Industries, Inc., Yokohama, Japan
| | | | - Takamasa Gomi
- From POLA Chemical Industries, Inc., Yokohama, Japan
| |
Collapse
|
4
|
Yu W, Wang Z, Dai Y, Zhao S, Chen H, Wang S, Xie H. Autologous fat grafting for postoperative breast reconstruction: A systemic review. Regen Ther 2024; 26:1010-1017. [PMID: 39553540 PMCID: PMC11564784 DOI: 10.1016/j.reth.2024.10.007] [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: 05/24/2024] [Revised: 09/22/2024] [Accepted: 10/10/2024] [Indexed: 11/19/2024] Open
Abstract
Autologous fat grafting technology has become a new method for breast reconstruction after breast surgery due to its advantages of simple operation, low immunogenicity, fewer complications, high patient acceptance, and natural filling effect. However, the unpredictable fate of transplanted fat limits its widespread application. Currently, many studies have made certain progress in improving the survival rate of fat grafts. This article provides an overview of autologous fat grafting technology, including the mechanisms of fat graft survival, techniques for obtaining and transplanting adipose tissue, methods for enhancing graft survival, and complications associated with fat grafting.
Collapse
Affiliation(s)
| | | | - Yuhan Dai
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Shuhan Zhao
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Huilin Chen
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Shui Wang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Hui Xie
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| |
Collapse
|
5
|
Zhang YA, Li FW, Dong YX, Xie WJ, Wang HB. PPAR-γ regulates the polarization of M2 macrophages to improve the microenvironment for autologous fat grafting. FASEB J 2024; 38:e23613. [PMID: 38661048 DOI: 10.1096/fj.202400126r] [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: 01/17/2024] [Revised: 03/19/2024] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
The unpredictable survival rate of autologous fat grafting (AFG) seriously affects its clinical application. Improving the survival rate of AFG has become an unresolved issue in plastic surgery. Peroxisome proliferator-activated receptor-γ (PPAR-γ) regulates the adipogenic differentiation of adipocytes, but the functional mechanism in AFG remains unclear. In this study, we established an animal model of AFG and demonstrated the superior therapeutic effect of PPAR-γ regulation in the process of AFG. From day 3 after fat grafting, the PPAR-γ agonist rosiglitazone group consistently showed better adipose integrity, fewer oil cysts, and fibrosis. Massive macrophage infiltration was observed after 7 days. At the same time, M2 macrophages begin to appear. At day 14, M2 macrophages gradually became the dominant cell population, which suppressed inflammation and promoted revascularization and fat regeneration. In addition, transcriptome sequencing showed that the differentially expressed genes in the Rosiglitazone group were associated with the pathways of adipose regeneration, differentiation, and angiogenesis; these results provide new ideas for clinical treatment.
Collapse
Affiliation(s)
- Ya-An Zhang
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Fang-Wei Li
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Yun-Xian Dong
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Wen-Jie Xie
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Hai-Bin Wang
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
6
|
Frommer ML, Langridge BJ, Beedie A, Jasionowska S, Awad L, Denton CP, Abraham DJ, Abu-Hanna J, Butler PEM. Exploring Anti-Fibrotic Effects of Adipose-Derived Stem Cells: Transcriptome Analysis upon Fibrotic, Inflammatory, and Hypoxic Conditioning. Cells 2024; 13:693. [PMID: 38667308 PMCID: PMC11049044 DOI: 10.3390/cells13080693] [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: 03/01/2024] [Revised: 04/08/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Autologous fat transfers show promise in treating fibrotic skin diseases, reversing scarring and stiffness, and improving quality of life. Adipose-derived stem cells (ADSCs) within these grafts are believed to be crucial for this effect, particularly their secreted factors, though the specific mechanisms remain unclear. This study investigates transcriptomic changes in ADSCs after in vitro fibrotic, inflammatory, and hypoxic conditioning. High-throughput gene expression assays were conducted on ADSCs exposed to IL1-β, TGF-β1, and hypoxia and in media with fetal bovine serum (FBS). Flow cytometry characterized the ADSCs. RNA-Seq analysis revealed distinct gene expression patterns between the conditions. FBS upregulated pathways were related to the cell cycle, replication, wound healing, and ossification. IL1-β induced immunomodulatory pathways, including granulocyte chemotaxis and cytokine production. TGF-β1 treatment upregulated wound healing and muscle tissue development pathways. Hypoxia led to the downregulation of mitochondria and cellular activity.
Collapse
Affiliation(s)
- Marvin L. Frommer
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK
- Department of Surgical Biotechnology, Division of Surgery & Interventional Science, University College London, London NW3 2QG, UK
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| | - Benjamin J. Langridge
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK
- Department of Surgical Biotechnology, Division of Surgery & Interventional Science, University College London, London NW3 2QG, UK
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| | - Alexandra Beedie
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| | - Sara Jasionowska
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| | - Laura Awad
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| | - Christopher P. Denton
- Centre for Rheumatology, Department of Inflammation and Rare Diseases, Division of Medicine, University College London, London NW3 2QG, UK
| | - David J. Abraham
- Centre for Rheumatology, Department of Inflammation and Rare Diseases, Division of Medicine, University College London, London NW3 2QG, UK
| | - Jeries Abu-Hanna
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK
- Division of Medical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Peter E. M. Butler
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK
- Department of Surgical Biotechnology, Division of Surgery & Interventional Science, University College London, London NW3 2QG, UK
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| |
Collapse
|
7
|
Hazrati A, Malekpour K, Khorramdelazad H, Rajaei S, Hashemi SM. Therapeutic and immunomodulatory potentials of mesenchymal stromal/stem cells and immune checkpoints related molecules. Biomark Res 2024; 12:35. [PMID: 38515166 PMCID: PMC10958918 DOI: 10.1186/s40364-024-00580-2] [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: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are used in many studies due to their therapeutic potential, including their differentiative ability and immunomodulatory properties. These cells perform their therapeutic functions by using various mechanisms, such as the production of anti-inflammatory cytokines, growth factors, direct cell-to-cell contact, extracellular vesicles (EVs) production, and mitochondrial transfer. However, mechanisms related to immune checkpoints (ICPs) and their effect on the immunomodulatory ability of MSCs are less discussed. The main function of ICPs is to prevent the initiation of unwanted responses and to regulate the immune system responses to maintain the homeostasis of these responses. ICPs are produced by various types of immune system regulatory cells, and defects in their expression and function may be associated with excessive responses that can ultimately lead to autoimmunity. Also, by expressing different types of ICPs and their ligands (ICPLs), tumor cells prevent the formation and durability of immune responses, which leads to tumors' immune escape. ICPs and ICPLs can be produced by MSCs and affect immune cell responses both through their secretion into the microenvironment or direct cell-to-cell interaction. Pre-treatment of MSCs in inflammatory conditions leads to an increase in their therapeutic potential. In addition to the effect that inflammatory environments have on the production of anti-inflammatory cytokines by MSCs, they can increase the expression of various types of ICPLs. In this review, we discuss different types of ICPLs and ICPs expressed by MSCs and their effect on their immunomodulatory and therapeutic potential.
Collapse
Affiliation(s)
- Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Samira Rajaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Pattani N, Sanghera J, Langridge BJ, Frommer ML, Abu-Hanna J, Butler P. Exploring the mechanisms behind autologous lipotransfer for radiation-induced fibrosis: A systematic review. PLoS One 2024; 19:e0292013. [PMID: 38271326 PMCID: PMC10810439 DOI: 10.1371/journal.pone.0292013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/11/2023] [Indexed: 01/27/2024] Open
Abstract
AIM Radiation-induced fibrosis is a recognised consequence of radiotherapy, especially after multiple and prolonged dosing regimens. There is no definitive treatment for late-stage radiation-induced fibrosis, although the use of autologous fat transfer has shown promise. However, the exact mechanisms by which this improves radiation-induced fibrosis remain poorly understood. We aim to explore existing literature on the effects of autologous fat transfer on both in-vitro and in-vivo radiation-induced fibrosis models, and to collate potential mechanisms of action. METHOD PubMed, Cochrane reviews and Scopus electronic databases from inception to May 2023 were searched. Our search strategy combined both free-text terms with Boolean operators, derived from synonyms of adipose tissue and radiation-induced fibrosis. RESULTS The search strategy produced 2909 articles. Of these, 90 underwent full-text review for eligibility, yielding 31 for final analysis. Nine conducted in-vitro experiments utilising a co-culture model, whilst 25 conducted in-vivo experiments. Interventions under autologous fat transfer included adipose-derived stem cells, stromal vascular function, whole fat and microfat. Notable findings include downregulation of fibroblast proliferation, collagen deposition, epithelial cell apoptosis, and proinflammatory processes. Autologous fat transfer suppressed hypoxia and pro-inflammatory interferon-γ signalling pathways, and tissue treated with adipose-derived stem cells stained strongly for anti-inflammatory M2 macrophages. Although largely proangiogenic initially, studies show varying effects on vascularisation. There is early evidence that adipose-derived stem cell subgroups may have different functional properties. CONCLUSION Autologous fat transfer functions through pro-angiogenic, anti-fibrotic, immunomodulatory, and extracellular matrix remodelling properties. By characterising these mechanisms, relevant drug targets can be identified and used to further improve clinical outcomes in radiation-induced fibrosis. Further research should focus on adipose-derived stem cell sub-populations and augmentation techniques such as cell-assisted lipotransfer.
Collapse
Affiliation(s)
| | | | - Benjamin J. Langridge
- Department of Plastic Surgery, Royal Free Hospital, London, United Kingdom
- Division of Surgery & Interventional Sciences, University College London, London, United Kingdom
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom
| | - Marvin L. Frommer
- Division of Surgery & Interventional Sciences, University College London, London, United Kingdom
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom
| | - Jeries Abu-Hanna
- Division of Surgery & Interventional Sciences, University College London, London, United Kingdom
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom
- Division of Medical Sciences, University of Oxford, Oxford, United Kingdom
| | - Peter Butler
- Department of Plastic Surgery, Royal Free Hospital, London, United Kingdom
- Division of Surgery & Interventional Sciences, University College London, London, United Kingdom
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
9
|
Isildar B, Ozkan S, Koyuturk M. Therapeutic Potential of Mesenchymal Stem Cell‐Derived Conditioned Medium for Diabetes Mellitus and Related Complications. ADVANCED THERAPEUTICS 2023; 6. [DOI: 10.1002/adtp.202300216] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Indexed: 01/06/2025]
Abstract
AbstractDiabetes mellitus (DM) is one of the most life‐threatening metabolic disorders, with 9% of the global prevalence, and it is estimated to be rising to 12.2% in 2045. Currently, there is no definitive treatment for DM. Although life‐saving, insulin administration to control blood sugar is not a cure for DM and is insufficient to prevent DM‐related complications such as nephropathy, neuropathy, or retinopathy. For this reason, studies are continuing to develop treatments that will provide β‐cell regeneration while suppressing autoimmunity. Mesenchymal stem cells (MSCs) are multipotent stem cells with a high proliferation capacity, immunosuppression, and immunomodulation ability. MSCs have gained therapeutic importance with these properties besides their differentiation ability. The immunosuppressive and immunomodulatory properties of the cells arise from the soluble and insoluble factors they secrete into the extracellular environment. Therefore, the culture medium where these cells grow has therapeutic value and is named conditioned medium (CM). In this context, CM obtained from MSCs can provide a similar therapeutic effect with fewer safety concerns. Furthermore, preconditioning of MSCs can improve the effectiveness of these cells and associated cellular products. So, this review summarizes the recent advances in MSC‐derived CMs and their therapeutic potential for DM and related complications.
Collapse
Affiliation(s)
- Basak Isildar
- Balikesir University Faculty of Medicine Histology and Embryology Department Balikesir 10185 Turkey
| | - Serbay Ozkan
- Izmir Katip Celebi University Faculty of Medicine Histology and Embryology Department Izmir 35620 Turkey
| | - Meral Koyuturk
- Istanbul University‐Cerrahpasa Cerrahpasa Faculty of Medicine Histology and Embryology Department Istanbul 34098 Turkey
| |
Collapse
|
10
|
Lin Y, Pan B, Mu D. Ferroptosis in Autologous Fat Transplantation: A New Hypothesis. J Craniofac Surg 2023; 34:e736-e739. [PMID: 37418612 DOI: 10.1097/scs.0000000000009508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 05/17/2023] [Indexed: 07/09/2023] Open
Abstract
Autologous adipose tissue transplantation is widely used for cosmetic and reconstruction of various areas in the body, often to repair soft tissue volume loss or contoured deformation. However, the application of fat transplantation is limited by unstable and unpredictable volume retention rates. At present, promoting adipose tissue survival and inhibiting its death is the key to improve the effect of autologous fat transplantation. In this paper, we propose a hypothesis that ferroptosis exists in fat transplantation. The bases of this hypothesis include the following: (1) the association between ferroptosis and other programmed cell death; (2) the association between ferroptosis and ischemia-reperfusion injury; and (3) the use of ferroptosis inhibitors in the field of fat transplantation.
Collapse
Affiliation(s)
- Yan Lin
- Department of Aesthetic and Reconstructive Breast Surgery
| | - Bo Pan
- Department of Auricular Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Dali Mu
- Department of Aesthetic and Reconstructive Breast Surgery
| |
Collapse
|
11
|
Jakl V, Popp T, Haupt J, Port M, Roesler R, Wiese S, Friemert B, Rojewski MT, Schrezenmeier H. Effect of Expansion Media on Functional Characteristics of Bone Marrow-Derived Mesenchymal Stromal Cells. Cells 2023; 12:2105. [PMID: 37626914 PMCID: PMC10453497 DOI: 10.3390/cells12162105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/07/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
The therapeutic efficacy of mesenchymal stromal cells (MSCs) has been shown to rely on their immunomodulatory and regenerative properties. In order to obtain sufficient numbers of cells for clinical applications, MSCs have to be expanded ex vivo. Expansion media with xenogeneic-free (XF) growth-promoting supplements like human platelet lysate (PL) or serum- and xenogeneic-free (SF/XF) formulations have been established as safe and efficient, and both groups provide different beneficial qualities. In this study, MSCs were expanded in XF or SF/XF media as well as in mixtures thereof. MSCs cultured in these media were analyzed for phenotypic and functional properties. MSC expansion was optimal with SF/XF conditions when PL was present. Metabolic patterns, consumption of growth factors, and secretome of MSCs differed depending on the type and concentration of supplement. The lactate per glucose yield increased along with a higher proportion of PL. Many factors in the supernatant of cultured MSCs showed distinct patterns depending on the supplement (e.g., FGF-2, TGFβ, and insulin only in PL-expanded MSC, and leptin, sCD40L PDGF-AA only in SF/XF-expanded MSC). This also resulted in changes in cell characteristics like migratory potential. These findings support current approaches where growth media may be utilized for priming MSCs for specific therapeutic applications.
Collapse
Affiliation(s)
- Viktoria Jakl
- Institute for Transfusion Medicine, University Hospital Ulm, 89081 Ulm, Germany; (V.J.)
| | - Tanja Popp
- Bundeswehr Institute of Radiobiology, 80937 Munich, Germany (J.H.); (M.P.)
| | - Julian Haupt
- Bundeswehr Institute of Radiobiology, 80937 Munich, Germany (J.H.); (M.P.)
- Clinic for Trauma Surgery and Orthopedics, Army Hospital Ulm, 89081 Ulm, Germany
| | - Matthias Port
- Bundeswehr Institute of Radiobiology, 80937 Munich, Germany (J.H.); (M.P.)
| | - Reinhild Roesler
- Core Unit of Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081 Ulm, Germany; (R.R.); (S.W.)
| | - Sebastian Wiese
- Core Unit of Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081 Ulm, Germany; (R.R.); (S.W.)
| | - Benedikt Friemert
- Clinic for Trauma Surgery and Orthopedics, Army Hospital Ulm, 89081 Ulm, Germany
| | - Markus T. Rojewski
- Institute for Transfusion Medicine, University Hospital Ulm, 89081 Ulm, Germany; (V.J.)
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg—Hessia and University Hospital Ulm, 89081 Ulm, Germany
| | - Hubert Schrezenmeier
- Institute for Transfusion Medicine, University Hospital Ulm, 89081 Ulm, Germany; (V.J.)
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg—Hessia and University Hospital Ulm, 89081 Ulm, Germany
| |
Collapse
|
12
|
Frommer ML, Langridge BJ, Awad L, Jasionowska S, Denton CP, Abraham DJ, Abu-Hanna J, Butler PEM. Single-Cell Analysis of ADSC Interactions with Fibroblasts and Endothelial Cells in Scleroderma Skin. Cells 2023; 12:1784. [PMID: 37443817 PMCID: PMC10341100 DOI: 10.3390/cells12131784] [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: 06/02/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) as part of autologous fat grafting have anti-fibrotic and anti-inflammatory effects, but the exact mechanisms of action remain unknown. By simulating the interaction of ADSCs with fibroblasts and endothelial cells (EC) from scleroderma (SSc) skin in silico, we aim to unravel these mechanisms. Publicly available single-cell RNA sequencing data from the stromal vascular fraction of 3 lean patients and biopsies from the skin of 10 control and 12 patients with SSc were obtained from the GEO and analysed using R and Seurat. Differentially expressed genes were used to compare the fibroblast and EC transcriptome between controls and SSc. GO and KEGG functional enrichment was performed. Ligand-receptor interactions of ADSCs with fibroblasts and ECs were explored with LIANA. Pro-inflammatory and extracellular matrix (ECM) interacting fibroblasts were identified in SSc. Arterial, capillary, venous and lymphatic ECs showed a pro-fibrotic and pro-inflammatory transcriptome. Most interactions with both cell types were based on ECM proteins. Differential interactions identified included NTN1, VEGFD, MMP2, FGF2, and FNDC5. The ADSC secretome may disrupt vascular and perivascular inflammation hubs in scleroderma by promoting angiogenesis and especially lymphangiogenesis. Key phenomena observed after fat grafting remain unexplained, including modulation of fibroblast behaviour.
Collapse
Affiliation(s)
- Marvin L. Frommer
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK; (B.J.L.); (J.A.-H.); (P.E.M.B.)
- Department of Surgical Biotechnology, Division of Surgery & Interventional Science, University College London, London NW3 2QG, UK
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| | - Benjamin J. Langridge
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK; (B.J.L.); (J.A.-H.); (P.E.M.B.)
- Department of Surgical Biotechnology, Division of Surgery & Interventional Science, University College London, London NW3 2QG, UK
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| | - Laura Awad
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK; (B.J.L.); (J.A.-H.); (P.E.M.B.)
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| | - Sara Jasionowska
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK; (B.J.L.); (J.A.-H.); (P.E.M.B.)
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| | - Christopher P. Denton
- Centre for Rheumatology, Department of Inflammation, Division of Medicine, University College London, London NW3 2QG, UK
| | - David J. Abraham
- Centre for Rheumatology, Department of Inflammation, Division of Medicine, University College London, London NW3 2QG, UK
| | - Jeries Abu-Hanna
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK; (B.J.L.); (J.A.-H.); (P.E.M.B.)
- Division of Medical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Peter E. M. Butler
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK; (B.J.L.); (J.A.-H.); (P.E.M.B.)
- Department of Surgical Biotechnology, Division of Surgery & Interventional Science, University College London, London NW3 2QG, UK
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| |
Collapse
|
13
|
Helissey C, Cavallero S, Guitard N, Théry H, Chargari C, François S. Revolutionizing Radiotoxicity Management with Mesenchymal Stem Cells and Their Derivatives: A Focus on Radiation-Induced Cystitis. Int J Mol Sci 2023; 24:ijms24109068. [PMID: 37240415 DOI: 10.3390/ijms24109068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/02/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Although radiation therapy plays a crucial role in cancer treatment, and techniques have improved continuously, irradiation induces side effects in healthy tissue. Radiation cystitis is a potential complication following the therapeutic irradiation of pelvic cancers and negatively impacts patients' quality of life (QoL). To date, no effective treatment is available, and this toxicity remains a therapeutic challenge. In recent times, stem cell-based therapy, particularly the use of mesenchymal stem cells (MSC), has gained attention in tissue repair and regeneration due to their easy accessibility and their ability to differentiate into several tissue types, modulate the immune system and secrete substances that help nearby cells grow and heal. In this review, we will summarize the pathophysiological mechanisms of radiation-induced injury to normal tissues, including radiation cystitis (RC). We will then discuss the therapeutic potential and limitations of MSCs and their derivatives, including packaged conditioned media and extracellular vesicles, in the management of radiotoxicity and RC.
Collapse
Affiliation(s)
- Carole Helissey
- Clinical Unit Research, HIA Bégin, 69 Avenu de Paris, 94160 Saint-Mandé, France
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, Place Général Valérie André, 91220 Brétigny-sur-Orge, France
| | - Sophie Cavallero
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, Place Général Valérie André, 91220 Brétigny-sur-Orge, France
| | - Nathalie Guitard
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, Place Général Valérie André, 91220 Brétigny-sur-Orge, France
| | - Hélène Théry
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, Place Général Valérie André, 91220 Brétigny-sur-Orge, France
| | - Cyrus Chargari
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, Place Général Valérie André, 91220 Brétigny-sur-Orge, France
- Department of Radiation Oncology, Pitié Salpêtrière University Hospital, 47-83 Bd de l'Hôpital, 75013 Paris, France
| | - Sabine François
- Department of Radiation Biological Effects, French Armed Forces Biomedical Research Institute, Place Général Valérie André, 91220 Brétigny-sur-Orge, France
| |
Collapse
|
14
|
Li P, Ou Q, Shi S, Shao C. Immunomodulatory properties of mesenchymal stem cells/dental stem cells and their therapeutic applications. Cell Mol Immunol 2023; 20:558-569. [PMID: 36973490 PMCID: PMC10040934 DOI: 10.1038/s41423-023-00998-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 03/02/2023] [Indexed: 03/29/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are widely distributed in the body and play essential roles in tissue regeneration and homeostasis. MSCs can be isolated from discarded tissues, expanded in vitro and used as therapeutics for autoimmune diseases and other chronic disorders. MSCs promote tissue regeneration and homeostasis by primarily acting on immune cells. At least six different types of MSCs have been isolated from postnatal dental tissues and have remarkable immunomodulatory properties. Dental stem cells (DSCs) have been demonstrated to have therapeutic effects on several systemic inflammatory diseases. Conversely, MSCs derived from nondental tissues such as the umbilical cord exhibit great benefits in the management of periodontitis in preclinical studies. Here, we discuss the main therapeutic uses of MSCs/DSCs, their mechanisms, extrinsic inflammatory cues and the intrinsic metabolic circuitries that govern the immunomodulatory functions of MSCs/DSCs. Increased understanding of the mechanisms underpinning the immunomodulatory functions of MSCs/DSCs is expected to aid in the development of more potent and precise MSC/DSC-based therapeutics.
Collapse
Affiliation(s)
- Peishan Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, PR China
| | - Qianmin Ou
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, PR China
| | - Songtao Shi
- South China Center of Craniofacial Stem Cell Research, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, PR China.
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, PR China.
| |
Collapse
|
15
|
Lagneau N, Tournier P, Nativel F, Maugars Y, Guicheux J, Le Visage C, Delplace V. Harnessing cell-material interactions to control stem cell secretion for osteoarthritis treatment. Biomaterials 2023; 296:122091. [PMID: 36947892 DOI: 10.1016/j.biomaterials.2023.122091] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/08/2023] [Accepted: 03/12/2023] [Indexed: 03/16/2023]
Abstract
Osteoarthritis (OA) is the most common debilitating joint disease, yet there is no curative treatment for OA to date. Delivering mesenchymal stromal cells (MSCs) as therapeutic cells to mitigate the inflammatory symptoms associated with OA is attracting increasing attention. In principle, MSCs could respond to the pro-inflammatory microenvironment of an OA joint by the secretion of anti-inflammatory, anti-apoptotic, immunomodulatory and pro-regenerative factors, therefore limiting pain, as well as the disease development. However, the microenvironment of MSCs is known to greatly affect their survival and bioactivity, and using tailored biomaterial scaffolds could be key to the success of intra-articular MSC-based therapies. The aim of this review is to identify and discuss essential characteristics of biomaterial scaffolds to best promote MSC secretory functions in the context of OA. First, a brief introduction to the OA physiopathology is provided, followed by an overview of the MSC secretory functions, as well as the current limitations of MSC-based therapy. Then, we review the current knowledge on the effects of cell-material interactions on MSC secretion. These considerations allow us to define rational guidelines for next-generation biomaterial design to improve the MSC-based therapy of OA.
Collapse
Affiliation(s)
- Nathan Lagneau
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, France
| | - Pierre Tournier
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, France
| | - Fabien Nativel
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, France; Nantes Université, UFR Sciences Biologiques et Pharmaceutiques, Nantes, F-44035, France
| | - Yves Maugars
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, France.
| | - Catherine Le Visage
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, France
| | - Vianney Delplace
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, France
| |
Collapse
|
16
|
Sun D, Mou S, Chen L, Yang J, Wang R, Zhong A, Wang W, Tong J, Wang Z, Sun J. High yield engineered nanovesicles from ADSC with enriched miR-21-5p promote angiogenesis in adipose tissue regeneration. Biomater Res 2022; 26:83. [PMID: 36528594 PMCID: PMC9758932 DOI: 10.1186/s40824-022-00325-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have been found to have a great potential for soft tissue repair due to various biological functions, including pro-angiogenesis and low immunogenicity. However, the low yield and heterogeneity of MSC-EVs limited their clinical transformation. This study was designed to develop a novel adipose-derived stem cell engineered nanovesicles (ADSC-NVs) with high production and explore its pro-angiogenetic effect and application in adipose tissue regeneration. METHODS Adipose-derived stem cell-derived extracellular vesicles (ADSC-EVs) were isolated from an EVs-free culture medium for human ADSCs (hADSCs). ADSC-NVs were prepared by sequentially extruding ADSCs followed by iodixanol density gradient ultracentrifugation and were compared with ADSC-EVs in morphology, size distribution, protein contents and yield. The pro-angiogenetic effect of ADSC-NVs in different doses (0, 5, 20 and 80 μg/mL) in vitro was determined using transwell assay, tube formation assay, western blot and qRT-PCR. In vivo, BALB/c nude mice were administered injection of a mixture of fat granules and different dose of ADSC-NVs and grafts were harvested at 12 weeks post-transplantation for further analysis. By analyzing the weight and volume of grafts and histological evaluation, we investigated the effect of ADSC-NVs in vessel formation and adipose tissue regeneration. RESULTS Our results showed yield of purified ADSC-NVs was approximately 20 times more than that of ADSC-EVs secreted by the same number of ADSCs. In vitro, both ADSC-NVs and ADSC-EVs exhibited a dose-dependent pro-angiogenetic effect, despite their distinct miRNA profiles. These effects of ADSC-NVs may be mediated by enriched miR-21-5p via PTEN inhibition and PI3K/p-Akt signaling activation. Furthermore, after a mixed injection of ADSC-NVs, vessel formation and adipose regeneration were observed in vivo in fat implants. CONCLUSIONS Our study developed a potent alternative of ADSC-EVs. ADSC-NVs have a high pro-angiogenesis potential and can be used as cell-free therapeutic biomaterials in soft tissue regeneration.
Collapse
Affiliation(s)
- Di Sun
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Shan Mou
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Lifeng Chen
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Jie Yang
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Rongrong Wang
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Aimei Zhong
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Wei Wang
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Jing Tong
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Zhenxing Wang
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Jiaming Sun
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| |
Collapse
|
17
|
Chun JJ, Chang J, Soedono S, Oh J, Kim YJ, Wee SY, Cho KW, Choi CY. Mechanical Stress Improves Fat Graft Survival by Promoting Adipose-Derived Stem Cells Proliferation. Int J Mol Sci 2022; 23:ijms231911839. [PMID: 36233141 PMCID: PMC9569524 DOI: 10.3390/ijms231911839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/28/2022] [Accepted: 10/01/2022] [Indexed: 11/16/2022] Open
Abstract
Cell-assisted lipotransfer (CAL), defined as co-transplantation of aspirated fat with enrichment of adipose-derived stem cells (ASCs), is a novel technique for cosmetic and reconstructive surgery to overcome the low survival rate of traditional fat grafting. However, clinically approved techniques for increasing the potency of ASCs in CAL have not been developed yet. As a more clinically applicable method, we used mechanical stress to reinforce the potency of ASCs. Mechanical stress was applied to the inguinal fat pad by needling. Morphological and cellular changes in adipose tissues were examined by flow cytometric analysis 1, 3, 5, and 7 days after the procedure. The proliferation and adipogenesis potencies of ASCs were evaluated. CAL with ASCs treated with mechanical stress or sham control were performed, and engraftment was determined at 4 weeks post-operation. Flow cytometry analysis revealed that mechanical stress significantly increased the number as well as the frequency of ASC proliferation in fat. Proliferation assays and adipocyte-specific marker gene analysis revealed that mechanical stress promoted proliferation potential but did not affect the differentiation capacity of ASCs. Moreover, CAL with cells derived from mechanical stress-treated fat increased the engraftment. Our results indicate that mechanical stress may be a simple method for improving the efficacy of CAL by enhancing the proliferation potency of ASCs.
Collapse
Affiliation(s)
- Jeong Jin Chun
- Department of Plastic and Reconstructive Surgery, Soonchunhyang University Hospital, Gumi 39371, Korea
| | - Jiyeon Chang
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Korea
| | - Shindy Soedono
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Korea
| | - Jieun Oh
- Department of Medical Biotechnology, Soonchunhyang University, Asan 31583, Korea
| | - Yeong Jin Kim
- Department of Plastic and Reconstructive Surgery, Soonchunhyang University Hospital, Bucheon 14584, Korea
| | - Syeo Young Wee
- Department of Plastic and Reconstructive Surgery, Soonchunhyang University Hospital, Gumi 39371, Korea
| | - Kae Won Cho
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Korea
- Correspondence: (K.W.C.); (C.Y.C.); Tel.: +82-41-413-5028 (K.W.C.); +82-32-621-5319 (C.Y.C.)
| | - Chang Yong Choi
- Department of Plastic and Reconstructive Surgery, Soonchunhyang University Hospital, Bucheon 14584, Korea
- Correspondence: (K.W.C.); (C.Y.C.); Tel.: +82-41-413-5028 (K.W.C.); +82-32-621-5319 (C.Y.C.)
| |
Collapse
|
18
|
Barisic G, Andjelkov K, Rosic J, Miladinov M, Kotur-Stеvuljevic J, Dinic T, Jelenkovic J, Krivokapic Z. Application of nanofat for treatment of traumatic faecal incontinence after sphincteroplasty - A pilot study. Colorectal Dis 2022; 24:1054-1062. [PMID: 35426481 DOI: 10.1111/codi.16148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/14/2022] [Accepted: 04/07/2022] [Indexed: 02/08/2023]
Abstract
AIM The aim of this study was to investigate whether the application of nanofat containing stem cells improves continence in women who had previously undergone anal sphincteroplasty with unsatisfactory long-term outcomes. METHOD This prospective pilot study included nine women with various degrees of anal incontinence who had previously undergone anal sphincteroplasty due to obstetric trauma. In all patients, the Wexner Incontinence Score (WS) and Faecal Incontinence Quality of Life Score (FIQLS), as well as anal manometry and endoanal ultrasound measurements, were performed before the procedure and during follow-up. In all patients, liposuction was performed and 50 ml of raw lipoaspirate was obtained and processed using a NanoFat Kit device. Approximately 20 ml of the mechanically emulsified and filtrated fat was obtained and the anal sphincter complex was infiltrated with it. Patient follow-up was conducted in person or via telephone 6 and 12 months after the procedure. RESULTS The squeeze pressure was significantly increased 6 months after the procedure (p = 0.01). The external anal sphincter measured at the 12 o'clock position was significantly thicker (p = 0.04). A significant decrease in the WS was observed both 6 and 12 months after the procedure compared with baseline values (p < 0.05 for both). CONCLUSION This study is the first to show that the application of nanofat as an injectable product improves continence in patients with unsatisfactory results after sphincteroplasty, suggesting it to be a promising and effective therapeutic tool. The procedure is safe and can be easily performed as an ambulatory procedure.
Collapse
Affiliation(s)
- Goran Barisic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia.,Clinic for Digestive Surgery - First Surgical Clinic, University Clinical Center of Serbia, Belgrade, Serbia
| | | | - Jovana Rosic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marko Miladinov
- Clinic for Digestive Surgery - First Surgical Clinic, University Clinical Center of Serbia, Belgrade, Serbia
| | | | - Tanja Dinic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelenko Jelenkovic
- COVID Hospital Batajnica, University Clinical Center of Serbia, Belgrade, Serbia
| | - Zoran Krivokapic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia.,Clinic for Digestive Surgery - First Surgical Clinic, University Clinical Center of Serbia, Belgrade, Serbia.,Serbian Academy of Sciences and Arts, Belgrade, Serbia
| |
Collapse
|
19
|
Tutak FN, Bakirhan EG. The effects of the recipient site on fat graft survival in a murine model. EUROPEAN JOURNAL OF PLASTIC SURGERY 2022. [DOI: 10.1007/s00238-022-01979-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
20
|
Han Y, Yang J, Fang J, Zhou Y, Candi E, Wang J, Hua D, Shao C, Shi Y. The secretion profile of mesenchymal stem cells and potential applications in treating human diseases. Signal Transduct Target Ther 2022; 7:92. [PMID: 35314676 PMCID: PMC8935608 DOI: 10.1038/s41392-022-00932-0] [Citation(s) in RCA: 300] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 11/18/2021] [Accepted: 02/20/2022] [Indexed: 02/06/2023] Open
Abstract
AbstractMesenchymal stromal/stem cells (MSCs) possess multi-lineage differentiation and self-renewal potentials. MSCs-based therapies have been widely utilized for the treatment of diverse inflammatory diseases, due to the potent immunoregulatory functions of MSCs. An increasing body of evidence indicates that MSCs exert their therapeutic effects largely through their paracrine actions. Growth factors, cytokines, chemokines, extracellular matrix components, and metabolic products were all found to be functional molecules of MSCs in various therapeutic paradigms. These secretory factors contribute to immune modulation, tissue remodeling, and cellular homeostasis during regeneration. In this review, we summarize and discuss recent advances in our understanding of the secretory behavior of MSCs and the intracellular communication that accounts for their potential in treating human diseases.
Collapse
|
21
|
Liu SY, Wu JJ, Chen ZH, Zou ML, Teng YY, Zhang KW, Li YY, Guo DY, Yuan FL, Li X. Insight into the role of dermal white adipose tissue loss in dermal fibrosis. J Cell Physiol 2021; 237:169-177. [PMID: 34608987 DOI: 10.1002/jcp.30552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/16/2022]
Abstract
The loss of dermal white adipose tissue (dWAT) is vital to the formation of dermal fibrosis (DF), but the specific mechanism is not well understood. A few studies are reviewed to explore the role of dWAT in the formation of DF. Recent findings indicated that the adipocytes-to-myofibroblasts transition in dWAT reflects the direct contribution to the DF formation. While adipose-derived stem cells (ADSCs) contained in dWAT express antifibrotic cytokines, the loss of ADSCs leads to skin protection decreased, which indirectly exacerbates DF and tissue damage. Therefore, blocking or reversing the adipocytes-to-myofibroblasts transition or improving the survival of ADSCs in dWAT and the expression of antifibrotic cytokines may be an effective strategy for the treatment of DF.
Collapse
Affiliation(s)
- Si-Yu Liu
- Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, Jiangsu, China
| | - Jun-Jie Wu
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu, China
| | - Zhong-Hua Chen
- Department of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Ming-Li Zou
- Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, Jiangsu, China
| | - Ying-Ying Teng
- Department of Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu, China
| | - Kai-Wen Zhang
- Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, Jiangsu, China
| | - Yue-Yue Li
- Department of Pharmacy, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu, China
| | - Dang-Yang Guo
- Department of Pharmacy, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu, China
| | - Feng-Lai Yuan
- Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, Jiangsu, China.,Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu, China.,Department of Pharmacy, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu, China
| | - Xia Li
- Department of Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
22
|
Chen A, Zhang L, Chen P, Zhang C, Tang S, Chen X. Comparison of the Efficacy and Safety of Cell-Assisted Lipotransfer and Platelet-Rich Plasma Assisted Lipotransfer: What Should We Expect from a Systematic Review with Meta-Analysis? Cell Transplant 2021; 30:963689721989607. [PMID: 33845642 PMCID: PMC8058798 DOI: 10.1177/0963689721989607] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Due to the high absorption rate of traditional autologous fat grafting, cell-assisted lipotransfer (CAL) and platelet-rich plasma (PRP)-assisted lipotransfer were developed. The purpose of this article was to evaluate the efficacy and safety of CAL and PRP in promoting the survival of autologous fat grafting through systematic review and meta-analysis. We searched Pubmed, Cochrane Library, Web of Science, and EMBASE for clinical studies on CAL and PRP-assisted lipotransfer published from January 2010 to January 2020. Then a meta-analysis was performed to assess the efficacy of CAL and PRP-assisted lipotransfer through data analysis of fat survival rate. We also assessed the incidence of complications and multiple operations to analyze their safety. A total of 36 studies (1697 patients) were included in this review. Regardless of the recipient area, CAL and PRP-assisted lipotransfer significantly improved the fat survival rate (CAL vs non-CAL: 71% vs 48%, P < 0.0001; PRP vs non-PRP: 70% vs 40%, P < 0.0001; CAL vs PRP: 71% vs 70%, P = 0.7175). However, in large-volume fat grafting, such as breast reconstruction, both increased the incidence of complications and did not decrease the frequency of multiple operations after lipotransfer. Further prospective studies are needed to evaluate the clinical benefits of CAL and PRP-assisted lipotransfer.
Collapse
Affiliation(s)
- Aizhen Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.,Both the authors contributed equally to this article and shared the first authorship
| | - Li Zhang
- Department of Central Sterile Services Department, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.,Both the authors contributed equally to this article and shared the first authorship
| | - Penghong Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chaoyu Zhang
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Shijie Tang
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Xiaosong Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
23
|
A novel method for processing adipose-derived stromal stem cells using a closed cell washing concentration device with a hollow fiber membrane module. Biomed Microdevices 2021; 23:3. [PMID: 33404966 PMCID: PMC7788025 DOI: 10.1007/s10544-020-00541-0] [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] [Accepted: 12/07/2020] [Indexed: 12/17/2022]
Abstract
Cell-assisted lipotransfer (CAL) is an advanced lipoinjection method that uses autologous lipotransfer with addition of a stromal vascular fraction (SVF) containing adipose-derived stromal stem cells (ASCs). The CAL procedure of manual isolation of cells from fat requires cell processing to be performed in clean environment. To isolate cells from fat without the need for a cell processing center, such as in a procedure in an operation theater, we developed a novel method for processing SVF using a closed cell washing concentration device (CCD) with a hollow fiber membrane module. The CCD consists of a sterilized closed circuit, bags and hollow fiber, semi-automatic device and the device allows removal of >99.97% of collagenase from SVF while maintaining sterility. The number of nucleated cells, ASCs and viability in SVF processed by this method were equivalent to those in SVF processed using conventional manual isolation. Our results suggest that the CCD system is as reliable as manual isolation and may also be useful for CAL. This approach will help in the development of regenerative medicine at clinics without a cell processing center.
Collapse
|
24
|
Horta R, Monteiro D, Órfão T, Nascimento R, Frias F, Silva A. Functional facial reconstruction in a patient with severe burn sequelae with a prelaminated parascapular free flap based on a three-dimensional model: A case report. Microsurgery 2020; 40:802-807. [PMID: 32961000 DOI: 10.1002/micr.30646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 05/04/2020] [Accepted: 08/21/2020] [Indexed: 11/05/2022]
Abstract
Significant evolution has been made concerning resuscitation and emergency management of severely burned patients, and nowadays most patients will survive and deal with burns sequelae. They constitute a reconstructive challenge, mainly because options and donor areas are frequently compromised, results are often limited, and other options should then be considered. A 27-year-old male patient with 55% total burn surface area, presented with severe facial disfigurement including ectropion, upper/lower lip retraction, and partial loss of the nose. In order to improve the patient's condition, autologous reconstruction was considered. The only unburned area in the body was the left dorsal region, and a three-stage reconstruction was planned using a paraescapular flap. In a first stage, an elective surgery was performed to identify and tag the recipient vessels in the neck. After 3 months, the prelamination process was initiated with the drawing of a facial model, and a nose and lips were opened inside the flap. This was based on a three-dimensional latex model as a print of the patient's face, which allowed us to calculate distances and estimate the length of the vascular pedicles. After 3 months, the flap (18 × 8 cm) was transferred and microvascular anastomoses were performed. No major complications were seen after surgeries, and after 28 months, an extremely important functional gain was obtained. Despite the number of surgeries required and less than optimal aesthetic results, this method may offer a satisfactory solution for complex acquired facial burn sequelae when other local or distant flap options are not available.
Collapse
Affiliation(s)
- Ricardo Horta
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar Universitário de São João, Faculty of Medicine - Porto University (FMUP), Porto, Portugal
| | - Diana Monteiro
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar Universitário de São João, Faculty of Medicine - Porto University (FMUP), Porto, Portugal
| | - Tiago Órfão
- Department of Otorhinolaryngology, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Ricardo Nascimento
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar Universitário de São João, Faculty of Medicine - Porto University (FMUP), Porto, Portugal
| | - Francisca Frias
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar Universitário de São João, Faculty of Medicine - Porto University (FMUP), Porto, Portugal
| | - Alvaro Silva
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar Universitário de São João, Faculty of Medicine - Porto University (FMUP), Porto, Portugal
| |
Collapse
|
25
|
Yang Z, Li H, Yuan Z, Fu L, Jiang S, Gao C, Wang F, Zha K, Tian G, Sun Z, Huang B, Wei F, Cao F, Sui X, Peng J, Lu S, Guo W, Liu S, Guo Q. Endogenous cell recruitment strategy for articular cartilage regeneration. Acta Biomater 2020; 114:31-52. [PMID: 32652223 DOI: 10.1016/j.actbio.2020.07.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/02/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023]
Abstract
In the absence of timely and proper treatments, injuries to articular cartilage (AC) can lead to cartilage degeneration and ultimately result in osteoarthritis. Regenerative medicine and tissue engineering techniques are emerging as promising approaches for AC regeneration and repair. Although the use of cell-seeded scaffolds prior to implantation can regenerate and repair cartilage lesions to some extent, these approaches are still restricted by limited cell sources, excessive costs, risks of disease transmission and complex manufacturing practices. Recently developed acellular scaffold approaches that rely on the recruitment of endogenous cells to the injured sites avoid these drawbacks and offer great promise for in situ AC regeneration. Multiple endogenous stem/progenitor cells (ESPCs) are found in joint-resident niches and have the capability to migrate to sites of injury to participate in AC regeneration. However, the natural recruitment of ESPCs is insufficient, and the local microenvironment is hostile after injury. Hence, an endogenous cell recruitment strategy based on the combination of chemoattractants and acellular scaffolds to effectively and specifically recruit ESPCs and improve local microenvironment may provide new insights into in situ AC regeneration. This review provides a brief overview of: (1) the status of endogenous cell recruitment strategy; (2) the subpopulations, potential migration routes (PMRs) of joint-resident ESPCs and their immunomodulatory and reparative effects; (3) chemoattractants and their potential adverse effects; (4) scaffold-based drug delivery systems (SDDSs) that are utilized for in situ AC regeneration; and (5) the challenges and future perspectives of endogenous cell recruitment strategy for AC regeneration. STATEMENT OF SIGNIFICANCE: Although the endogenous cell recruitment strategy for articular cartilage (AC) regeneration has been investigated for several decades, much work remains to be performed in this field. Future studies should have the following aims: (1) reporting the up-to-date progress in the endogenous cell recruitment strategies; (2) determining the subpopulations of ESPCs, the cellular and molecular mechanisms underlying the migration of these cells and their anti-inflammatory, immunomodulatory and reparative effects; (3) elucidating the chemoattractants that enhance ESPC recruitment and their potential adverse effects; and (4) developing advanced SDDSs for chemoattractant dispatch. Herein, we present a systematic overview of the aforementioned issues to provide a better understanding of endogenous cell recruitment strategies for AC regeneration and repair.
Collapse
|
26
|
Ma J, Yan X, Lin Y, Tan Q. Hepatocyte Growth Factor Secreted from Human Adipose-Derived Stem Cells Inhibits Fibrosis in Hypertrophic Scar Fibroblasts. Curr Mol Med 2020; 20:558-571. [PMID: 31903876 DOI: 10.2174/1566524020666200106095745] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 11/22/2022]
Abstract
AIMS To study the effect of Adipose-derived stem cells (ADSCs) on fibrosis of hypertrophic scar-derived fibroblasts (HSFs) and its concrete mechanism. BACKGROUND ADSCs have been reported to reduce collagen production and fibroblast proliferation in co-culture experiments. Conditioned medium from adipose-derived stem cells (ADSCs-CM) has successfully inhibited fibrosis by decreasing the expression of collagen type І (Col1) and α-smooth muscle actin (α-SMA) in rabbit ear scar models. Hepatocyte growth factor (HGF), the primary growth factor in ADSCs-CM, has been shown to reverse fibrosis in various fibrotic diseases. OBJECTIVE To test the hypothesis that ADSCs inhibit fibrosis of HSFs through the secretion of HGF. METHODS HSFs were treated with DMEM containing 0%, 10%, 50% and 100% concentration of ADSCs-CM. The effect of ADSCs-CM on the viability was determined by cell viability assay, and the collagen production in HSFs was examined by Sirius red staining. Expression and secretion of fibrosis and degradation proteins were detected separately. After measuring the concentration of HGF in ADSCs-CM, the same number of HSFs were treated with 50% ADSCs-CM or HGF. HGF activity in ADSCs-CM was neutralized with a goat anti-human HGF antibody. RESULTS The results demonstrated that ADSCs-CM dose-dependently decreased cell viability, expression of fibrosis molecules, and tissue inhibitor of metalloproteinases-1 (TIMP-1), and significantly increased matrix metalloproteinase-1 (MMP-1) expression in HSFs. Collagen production and the ratio of collagen type І and type III (Col1/Col3) were also suppressed by ADSCs-CM in a dose-dependent manner. When HSFs were cultured with either 50% ADSCs-CM or HGF (1 ng/ml), a similar trend was observed in gene expression and protein secretion. Adding an HGF antibody to both groups returned protein expression and secretion to basal levels but did not significantly affect the fibrosis factors in the control group. CONCLUSION Our findings revealed that adipose-derived stem cell-secreted HGF effectively inhibits fibrosis-related factors and regulates extracellular matrix (ECM) remodeling in hypertrophic scar fibroblasts.
Collapse
Affiliation(s)
- Ji Ma
- 1Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210008, China
| | - Xin Yan
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Yue Lin
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Qian Tan
- 1Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210008, China
| |
Collapse
|
27
|
Hara T, Soyama A, Adachi T, Kobayashi S, Sakai Y, Maruya Y, Kugiyama T, Hidaka M, Okada S, Hamada T, Maekawa K, Ono S, Adachi T, Takatsuki M, Eguchi S. Ameliorated healing of biliary anastomosis by autologous adipose-derived stem cell sheets. Regen Ther 2020; 14:79-86. [PMID: 31988997 PMCID: PMC6970135 DOI: 10.1016/j.reth.2019.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/10/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Cell sheets consisting of adipose-derived stem cells (ADSCs) have been reported to be effective for wound healing. We conducted this study to clarify the efficacy of ADSC sheets in wound healing at the duct-to-duct biliary anastomotic site in pigs. METHODS Eleven female pigs (20-25 kg) were divided into two groups: biliary anastomosis with an ADSC sheet (n = 6) or without an ADSC sheet (n = 5). To follow the transplanted ADSCs, PKH26GL-labeled sheets were used in one of the ADSC pigs. Two weeks prior to laparotomy, ADSCs were isolated from the lower abdominal subcutaneous adipose tissue. After three passages, ADSCs were seeded on temperature-responsive culture dishes and collected as cell sheets. ADSC sheets were gently transplanted on the anastomotic site. We evaluated specimens by PKH26GL labeling, macroscopic changes, infiltration of inflammatory cells, and collagen content. RESULTS Labeled ADSCs remained around the bile duct wall. In the no-ADSC group, more adhesion developed at the hepatic hilum as observed during relaparotomy. Histopathological examination showed that the diameter and cross-sectional area of the bile duct wall were decreased in the ADSC group. In the no-ADSC group, a large number of inflammatory cells and more collagen fibers were identified in the bile duct wall. CONCLUSIONS The present study demonstrated that autologous ADSC sheet transplantation reduced hypertrophic changes in the bile duct wall at the anastomotic site. A long-term follow-up is required to evaluate the efficacy of this mechanism in prevention of biliary anastomotic strictures.
Collapse
Key Words
- ADSC, adipose-derived stem cell
- APC, allophycocyanin
- Adipose-derived stem cell
- Anastomotic healing
- BAS, biliary anastomotic strictures
- BMSC, bone marrow stem cells
- Biliary anastomosis
- CBD, common bile duct
- Cell sheet
- FBS, fetal bovine serum
- FGF, fibroblast growth factor
- HGF, hepatocyte growth factor
- MSC, mesenchymal stem cell
- VEGF, vascular endothelial growth factor
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Susumu Eguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| |
Collapse
|
28
|
Conci C, Bennati L, Bregoli C, Buccino F, Danielli F, Gallan M, Gjini E, Raimondi MT. Tissue engineering and regenerative medicine strategies for the female breast. J Tissue Eng Regen Med 2019; 14:369-387. [PMID: 31825164 PMCID: PMC7065113 DOI: 10.1002/term.2999] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 11/06/2019] [Accepted: 11/11/2019] [Indexed: 12/16/2022]
Abstract
The complexity of mammary tissue and the variety of cells involved make tissue regeneration an ambitious goal. This review, supported by both detailed macro and micro anatomy, illustrates the potential of regenerative medicine in terms of mammary gland reconstruction to restore breast physiology and morphology, damaged by mastectomy. Despite the widespread use of conventional therapies, many critical issues have been solved using the potential of stem cells resident in adipose tissue, leading to commercial products. in vitro research has reported that adipose stem cells are the principal cellular source for reconstructing adipose tissue, ductal epithelium, and nipple structures. In addition to simple cell injection, construct made by cells seeded on a suitable biodegradable scaffold is a viable alternative from a long‐term perspective. Preclinical studies on mice and clinical studies, most of which have reached Phase II, are essential in the commercialization of cellular therapy products. Recent studies have revealed that the enrichment of fat grafting with stromal vascular fraction cells is a viable alternative to breast reconstruction. Although in the future, organ‐on‐a‐chip can be envisioned, for the moment researchers are still focusing on therapies that are a long way from regenerating the whole organ, but which nevertheless prevent complications, such as relapse and loss in terms of morphology.
Collapse
Affiliation(s)
- Claudio Conci
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Lorenzo Bennati
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Chiara Bregoli
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Federica Buccino
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Francesca Danielli
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Michela Gallan
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Ereza Gjini
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| | - Manuela T Raimondi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| |
Collapse
|
29
|
Ejaz A, Greenberger JS, Rubin PJ. Understanding the mechanism of radiation induced fibrosis and therapy options. Pharmacol Ther 2019; 204:107399. [DOI: 10.1016/j.pharmthera.2019.107399] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023]
|
30
|
Rode MP, Batti Angulski AB, Gomes FA, da Silva MM, Jeremias TDS, de Carvalho RG, Iucif Vieira DG, Oliveira LFC, Fernandes Maia L, Trentin AG, Hayashi L, de Miranda KR, de Aguiar AK, Rosa RD, Calloni GW. Carrageenan hydrogel as a scaffold for skin-derived multipotent stromal cells delivery. J Biomater Appl 2019; 33:422-434. [PMID: 30223731 DOI: 10.1177/0885328218795569] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Carrageenan is a thermoreversible polymer of natural origin widely used in food and pharmaceutical industry that presents a glycosaminoglycan-like structure. Herein, we show that kappa-type carrageenan extracted by a semi-refined process from the red seaweed Kappaphycus alvarezii displayed both chemical and structural properties similar to a commercial carrageenan. Moreover, both extracted carrageenan hydrogel and commercial carrageenan hydrogel can serve as a scaffold for in vitro culture of human skin-derived multipotent stromal cells, demonstrating considerable potential as cell-carrier materials for cell delivery in tissue engineering. Skin-derived multipotent stromal cells cultured inside the carrageenan hydrogels showed a round shape morphology and maintained their growth and viability for at least one week in culture. Next, the effect of the extracted carrageenan hydrogel loaded with human skin-derived multipotent stromal cells was evaluated in a mouse model of full-thickness skin wound. Macroscopic and histological analyses revealed some pointed ameliorated features, such as reduced inflammatory process, faster initial recovery of wounded area, and improved extracellular matrix deposition. These results indicate that extracted carrageenan hydrogel can serve as a scaffold for in vitro growth and maintenance of human SD-MSCs, being also able to act as a delivery system of cells to wounded skin. Thus, evaluation of the properties discussed in this study contribute to a further understanding and specificities of the potential use of carrageenan hydrogel as a delivery system for several applications, further to skin wound healing.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Leila Hayashi
- Universidade Federal de Santa Catarina, Florianopolis, Brazil
| | | | | | | | | |
Collapse
|
31
|
Vu NB, Phi LT, Dao TTT, Le HTN, Ta VT, Pham PV. Adipose derived stem cell transplantation is better than bone marrow mesenchymal stem cell transplantation in treating hindlimb ischemia in mice. BIOMEDICAL RESEARCH AND THERAPY 2019. [DOI: 10.7603/s40730-016-0046-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
32
|
The Effects of Adipose Stem Cell-Conditioned Media on Fibrogenesis of Dermal Fibroblasts Stimulated by Transforming Growth Factor-β1. J Burn Care Res 2019; 39:129-140. [PMID: 29931303 PMCID: PMC6083853 DOI: 10.1097/bcr.0000000000000558] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 03/05/2018] [Indexed: 01/09/2023]
Abstract
Adipose-derived stem cells (ASCs) have been shown to enhance wound healing by human dermal fibroblasts; however, the interactions between ASCs and fibroblasts during injury remain unclear. Fibroblasts were treated with ASC-conditioned medium (ASC-CM) with and without transforming growth factor-β1 (TGF-β1) stimulation. Fibroblast proliferation, apoptosis, differentiation and expression of extracellular matrix genes and proteins, type I collagen, and type III collagen were measured. Also, wound-healing effect of ASC-CM was verified with in vivo animal study. ASC-CM inhibited proliferation and enhanced apoptosis of fibroblasts under TGF-β1 stimulation. Furthermore, 10% ASC-CM inhibited α-smooth muscle actin expression in fibroblasts, whereas 100% ASC-CM increased collagen, especially type III, expression in fibroblasts. ASC-CM was found to contain more basic fibroblast growth factor than hepatocyte growth factor, and 100% ASC-CM increased hepatocyte growth factor gene expression in fibroblasts. These results suggest ASCs affect fibrogenesis by dermal fibroblasts stimulated with TGF-β1 via paracrine signaling by adipocytokines present in ASC-CM. These results also suggest that higher concentrations of ASC-CM increase collagen production and inhibit fibroblast proliferation to avoid excessive fibrogenesis. We demonstrated that a lower ASC-CM concentration attenuated fibroblast differentiation. Additionally, 100% ASC-CM significantly reduced the wound size in an in vivo wound-healing model. In this study, we provided evidence that ASCs modulate fibrogenesis by fibroblasts via paracrine signaling, suggesting that application of ASCs during wound healing may improve the quality of wound repair.
Collapse
|
33
|
Adipose-Derived Tissue in the Treatment of Dermal Fibrosis: Antifibrotic Effects of Adipose-Derived Stem Cells. Ann Plast Surg 2019; 80:297-307. [PMID: 29309331 DOI: 10.1097/sap.0000000000001278] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Treatment of hypertrophic scars and other fibrotic skin conditions with autologous fat injections shows promising clinical results; however, the underlying mechanisms of its antifibrotic action have not been comprehensively studied. Adipose-derived stem cells, or stromal cell-derived factors, inherent components of the transplanted fat tissue, seem to be responsible for its therapeutic effects on difficult scars. The mechanisms by which this therapeutic effect takes place are diverse and are mostly mediated by paracrine signaling, which switches on various antifibrotic molecular pathways, modulates the activity of the central profibrotic transforming growth factor β/Smad pathway, and normalizes functioning of fibroblasts and keratinocytes in the recipient site. Direct cell-to-cell communications and differentiation of cell types may also play a positive role in scar treatment, even though they have not been extensively studied in this context. A more thorough understanding of the fat tissue antifibrotic mechanisms of action will turn this treatment from an anecdotal remedy to a more controlled, timely administered technology.
Collapse
|
34
|
Ejaz A, Epperly MW, Hou W, Greenberger JS, Rubin JP. Adipose-Derived Stem Cell Therapy Ameliorates Ionizing Irradiation Fibrosis via Hepatocyte Growth Factor-Mediated Transforming Growth Factor-β Downregulation and Recruitment of Bone Marrow Cells. Stem Cells 2019; 37:791-802. [PMID: 30861238 DOI: 10.1002/stem.3000] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 02/06/2019] [Accepted: 02/22/2019] [Indexed: 01/10/2023]
Abstract
Radiation therapy to anatomic regions, including the head and neck, chest wall, and extremities, can produce radiation-induced fibrosis (RIF). To elucidate the cellular and molecular mechanism(s) involved in RIF, female C57BL/6J mice were irradiated to the right flank to 35 Gy in single fraction using 6 Mv electrons. Radiation fibrosis was detected by day 14, was increased by day 28, and confirmed by Masson's trichrome histological staining for collagen. Biopsied tissue at day 14 showed an increase in expression of fibrosis-related genes including transforming growth factor-β (TGF-β) and collagens 1-6. A single adipose-derived stem cell (ASC) injection on day 28 at the irradiated site decreased by day 40: epithelial thickness, collagen deposition, and significantly improved limb excursion compared with irradiated controls. Noncontact transwell coculture of ASCs above a monolayer of irradiated human foreskin fibroblasts downregulated fibrosis-related genes TGF-β, connective tissue growth factor, interleukin-1, NF-kB, tumor necrosis factor, and collagens 1-6. Hepatocyte growth factor (HGF) secreted by ASCs was identified as a novel mechanism by which ASCs exert antifibrotic effects by downregulating fibrotic gene expression in irradiated cells and recruiting bone marrow cells to the irradiated site. In conclusion, these data indicate a mechanistic role of HGF secreted by ASCs in reducing RIF. Stem Cells 2019;37:791-802.
Collapse
Affiliation(s)
- Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Michael W Epperly
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Wen Hou
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Joel S Greenberger
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - J Peter Rubin
- Department of Plastic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
35
|
Assisting Rapid Soft-Tissue Expansion with Adipose-Derived Stem Cells: An Experimental Study in a Pig Model. Plast Reconstr Surg 2019; 142:674e-684e. [PMID: 30511978 DOI: 10.1097/prs.0000000000004884] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Rapid tissue expansion has been attempted, aiming at shortening the period of conventional expansion. However, it has scarcely been clinically applied because of its drawbacks such as low expansion efficiency and tissue destruction. Adipose-derived stem cell transplantation is a promising therapeutic method in regenerative medicine. However, its effects on rapid expansion remain poorly understood. METHODS Twenty-four expanders were implanted in the dorsum of 12 pigs. Rapid expansion persisted for 1 week with 20 ml of saline daily. The increased area of the expanded skin was measured. Histologic and ultrastructural analysis and cell tracking were performed. The expression of vascular endothelial growth factor, fibroblast growth factor-2, and epidermal growth factor was also determined. RESULTS The increased area of adipose-derived stem cell-grafted expanded skin (0.91 ± 0.06 cm) was significantly more than the non-adipose-derived stem cell-treated control (0.51 ± 0.05 cm) (p < 0.01). Enhanced tissue regeneration in the adipose-derived stem cell-grafted expanded skin was evidenced by increased skin thickness, proliferating cells, extracellular matrix, and vascularization (113 ± 19/mm versus control 59 ± 14/mm) (all p < 0.05). Higher expression of vascular endothelial growth factor and epidermal growth factor was observed in the adipose-derived stem cell-transplanted expanded skin (p < 0.01 and p < 0.05, respectively), whereas the expression of fibroblast growth factor-2 was higher in the non-adipose-derived stem cell-treated control (p < 0.05). Transmission electron microscopy showed that a high density of collagen fibers could be seen in the adipose-derived stem cell-treated expanded skin. Cell tracking showed that the positively stained cells could be seen. CONCLUSION For rapid tissue expansion, adipose-derived stem cell transplantation may limit tissue destruction and improve the expansion efficiency by promoting tissue regeneration.
Collapse
|
36
|
Mancuso P, Raman S, Glynn A, Barry F, Murphy JM. Mesenchymal Stem Cell Therapy for Osteoarthritis: The Critical Role of the Cell Secretome. Front Bioeng Biotechnol 2019; 7:9. [PMID: 30761298 PMCID: PMC6361779 DOI: 10.3389/fbioe.2019.00009] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 01/11/2019] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is an inflammatory condition still lacking effective treatments. Mesenchymal stem/stromal cells (MSCs) have been successfully employed in pre-clinical models aiming to resurface the degenerated cartilage. In early-phase clinical trials, intra-articular (IA) administration of MSCs leads to pain reduction and cartilage protection or healing. However, the consistent lack of engraftment indicates that the observed effect is delivered through a "hit-and-run" mechanism, by a temporal release of paracrine molecules. MSCs express a variety of chemokines and cytokines that aid in repair of degraded tissue, restoration of normal tissue metabolism and, most importantly, counteracting inflammation. Secretion of therapeutic factors is increased upon licensing by inflammatory signals or apoptosis, induced by the host immune system. Trophic effectors are released as soluble molecules or carried by extracellular vesicles (ECVs). This review provides an overview of the functions and mechanisms of MSC-secreted molecules found to be upregulated in models of OA, whether using in vitro or in vivo models.
Collapse
Affiliation(s)
- Patrizio Mancuso
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), Biosciences, National University of Ireland Galway, Galway, Ireland
| | - Swarna Raman
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland
| | - Aoife Glynn
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland
| | - Frank Barry
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), Biosciences, National University of Ireland Galway, Galway, Ireland
| | - J Mary Murphy
- Regenerative Medicine Institute (REMEDI), Biosciences, National University of Ireland Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), Biosciences, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
37
|
Koledova Z, Sumbal J. FGF signaling in mammary gland fibroblasts regulates multiple fibroblast functions and mammary epithelial morphogenesis. Development 2019; 146:dev.185306. [DOI: 10.1242/dev.185306] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 10/24/2019] [Indexed: 12/20/2022]
Abstract
Fibroblast growth factor (FGF) signaling is crucial for mammary gland development. While multiple roles for FGF signaling in the epithelium were described, the function of FGF signaling in mammary stroma has not been elucidated. In this study, we investigated FGF signaling in mammary fibroblasts. We found that mammary fibroblasts express FGF receptors FGFR1 and FGFR2 and respond to FGF ligands. In particular, FGF2 and FGF9 induce sustained ERK1/2 signaling and promote fibroblast proliferation and migration in 2D. Intriguingly, only FGF2 induces fibroblast migration in 3D extracellular matrix (ECM) through regulation of actomyosin cytoskeleton and promotes force-mediated collagen remodeling by mammary fibroblasts. Moreover, FGF2 regulates production of ECM proteins by mammary fibroblasts, including collagens, fibronectin, osteopontin, and matrix metalloproteinases. Finally, using organotypic 3D co-cultures we show that FGF2 and FGF9 signaling in mammary fibroblasts enhances fibroblast-induced branching of mammary epithelium by modulating paracrine signaling and that knockdown of Fgfr1 and Fgfr2 in mammary fibroblasts reduces branching of mammary epithelium. Our results demonstrate a pleiotropic role for FGF signaling in mammary fibroblasts with implications for regulation of mammary stromal functions and epithelial branching morphogenesis.
Collapse
Affiliation(s)
- Zuzana Koledova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, Brno, 625 00, Czech Republic
| | - Jakub Sumbal
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, Brno, 625 00, Czech Republic
| |
Collapse
|
38
|
Chen J, Zhou Z, Yao Y, Dai J, Zhou D, Wang L, Zhang Q. Dipalmitoylphosphatidic acid inhibits breast cancer growth by suppressing angiogenesis via inhibition of the CUX1/FGF1/HGF signalling pathway. J Cell Mol Med 2018; 22:4760-4770. [PMID: 30010249 PMCID: PMC6156235 DOI: 10.1111/jcmm.13727] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/19/2018] [Indexed: 12/22/2022] Open
Abstract
Tumour growth depends on a continual supply of the nutrients and oxygen, which are offered by tumour angiogenesis. Our previous study showed that dipalmitoylphosphatidic acid (DPPA), a bioactive phospholipid, inhibits the growth of triple-negative breast cancer cells. However, its direct effect on angiogenesis remains unknown. Our work showed that DPPA significantly suppressed vascular growth in the chick embryo chorioallantoic membrane (CAM) and yolk sac membrane (YSM) models. Meanwhile, tumour angiogenesis and tumour growth were inhibited by DPPA in the tumour tissues of an experimental breast cancer model, a subcutaneous xenograft mouse model and a genetically engineered spontaneous breast cancer mouse model (MMTV-PyMT). Furthermore, DPPA directly inhibited the proliferation, migration and tube formation of vascular endothelial cells. The anti-angiogenic effect of DPPA was regulated by the inhibition of Cut-like homeobox1 (CUX1), which transcriptionally inhibited fibroblast growth factor 1 (FGF1), leading to the downregulation of hepatocyte growth factor (HGF). This work first demonstrates that DPPA directly inhibits angiogenesis in cancer development. Our previous work along with this study suggest that DPPA functions as an anti-tumour therapeutic drug that inhibits angiogenesis.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Chick Embryo
- Chorioallantoic Membrane/blood supply
- Chorioallantoic Membrane/drug effects
- Female
- Fibroblast Growth Factor 1/genetics
- Fibroblast Growth Factor 1/metabolism
- Gene Expression Regulation, Neoplastic
- Hepatocyte Growth Factor/genetics
- Hepatocyte Growth Factor/metabolism
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Human Umbilical Vein Endothelial Cells/cytology
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Nude
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Phosphatidic Acids/pharmacology
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction/drug effects
- Transcription Factors
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/pathology
- Tumor Burden/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jian Chen
- Vascular Biology Research InstituteSchool of Basic CourseGuangdong Pharmaceutical UniversityGuangzhouChina
| | - Zijun Zhou
- Vascular Biology Research InstituteSchool of Basic CourseGuangdong Pharmaceutical UniversityGuangzhouChina
| | - Yuying Yao
- Vascular Biology Research InstituteSchool of Basic CourseGuangdong Pharmaceutical UniversityGuangzhouChina
| | - Jianwei Dai
- GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouChina
- The State Key Lab of Respiratory DiseaseGuangzhou Institute of Respiratory DiseaseThe First Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Dalei Zhou
- Vascular Biology Research InstituteSchool of Basic CourseGuangdong Pharmaceutical UniversityGuangzhouChina
| | - Lijing Wang
- Vascular Biology Research InstituteSchool of Basic CourseGuangdong Pharmaceutical UniversityGuangzhouChina
| | - Qian‐Qian Zhang
- Vascular Biology Research InstituteSchool of Basic CourseGuangdong Pharmaceutical UniversityGuangzhouChina
| |
Collapse
|
39
|
Fukuoka H, Narita K, Suga H. Hair Regeneration Therapy: Application of Adipose-Derived Stem Cells. Curr Stem Cell Res Ther 2018; 12:531-534. [PMID: 28530535 PMCID: PMC6327114 DOI: 10.2174/1574888x12666170522114307] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 02/12/2017] [Accepted: 04/22/2017] [Indexed: 12/24/2022]
Abstract
BACKGROUND Adipose-derived stem cells secrete various cytokines that promote hair growth. OBJECTIVE To describe our experience of hair regeneration therapy using adipose-derived stem cellconditioned medium. RESULTS We performed the hair regeneration therapy in numerous Japanese patients and reported good results. We described characteristics of the commercialized conditioned medium, treatment methods, and future directions. CONCLUSION Treatment using adipose-derived stem cell-conditioned medium is highly effective and may represent a new therapy for alopecia.
Collapse
Affiliation(s)
| | - Keigo Narita
- Department of Plastic Surgery, Kyorin University School of Medicine, Tokyo. Japan
| | - Hirotaka Suga
- Department of Plastic Surgery, Kyorin University School of Medicine, Tokyo. Japan
| |
Collapse
|
40
|
Bi HS, Zhang C, Nie FF, Pan BL, Xiao E. Basic and Clinical Evidence of an Alternative Method to Produce Vivo Nanofat. Chin Med J (Engl) 2018; 131:588-593. [PMID: 29483394 PMCID: PMC5850676 DOI: 10.4103/0366-6999.226074] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Fat grafting technologies are popularly used in plastic and reconstructive surgery. Due to its size limitation, it is hard to directly inject untreated fat tissue into the dermal layer. Nanofat, which was introduced by Tonnard, solves this problem by mechanically emulsifying fat tissue. However, the viability of the cells was greatly destroyed. In this study, we reported a new method by “gently” digesting the fat tissue to produce viable adipocytes, progenitors, and stromal stem cells using collagenase I digestion and centrifugation. This was named “Vivo nanofat”. Methods: Human liposuction aspirates were obtained from five healthy female donors with mean age of 28.7 ± 5.6 years. Colony-forming assay, flow cytometry analysis, and adipogenic and osteogenic induction of the adherent cells from the Vivo nanofat were used to characterize the adipose mesenchymal stem cells (MSCs). To investigate in vivo survival, we respectively injected Vivo nanofat and nanofat subcutaneously to the back of 8-week-old male BALB/c nude mice. Samples were harvested 2 days, 2 weeks, and 4 weeks postinjection for measurement, hematoxylin and eosin staining, and immunostaining. Results: Our results showed that the Vivo nanofat contained a large number of colony-forming cells. These cells expressed MSC markers and had multi-differentiative potential. In vivo transplantation showed that the Vivo nanofat had lower resorption ratio than that of nanofat. The size of the transplanted nanofat was obviously smaller than that of Vivo nanofat 4 weeks postinjection (0.50 ± 0.17 cm vs. 0.81 ± 0.07 cm, t = −5783, P = 0.01). Conclusion: Vivo nanofat may serve as a cell fraction injectable through a fine needle; this could be used for cosmetic applications.
Collapse
Affiliation(s)
- Hong-Sen Bi
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Chen Zhang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Fang-Fei Nie
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Bo-Lin Pan
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - E Xiao
- Department of Oral and Maxillofacial Surgery, Peking University, School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| |
Collapse
|
41
|
Mattiucci D, Maurizi G, Leoni P, Poloni A. Aging- and Senescence-associated Changes of Mesenchymal Stromal Cells in Myelodysplastic Syndromes. Cell Transplant 2018; 27:754-764. [PMID: 29682980 PMCID: PMC6047275 DOI: 10.1177/0963689717745890] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Hematopoietic stem and progenitor cells reside within the bone marrow (BM) microenvironment. By a well-balanced interplay between self-renewal and differentiation, they ensure a lifelong supply of mature blood cells. Physiologically, multiple different cell types contribute to the regulation of stem and progenitor cells in the BM microenvironment by cell-extrinsic and cell-intrinsic mechanisms. During the last decades, mesenchymal stromal cells (MSCs) have been identified as one of the main cellular components of the BM microenvironment holding an indispensable role for normal hematopoiesis. During aging, MSCs diminish their functional and regenerative capacities and in some cases encounter replicative senescence, promoting inflammation and cancer progression. It is now evident that alterations in specific stromal cells that comprise the BM microenvironment can contribute to hematologic malignancies, and there is growing interest regarding the contribution of MSCs to the pathogenesis of myelodysplastic syndromes (MDSs), a clonal hematological disorder, occurring mostly in the elderly, characterized by ineffective hematopoiesis and increased tendency to acute myeloid leukemia evolution. The pathogenesis of MDS has been associated with specific genetic and epigenetic events occurring both in hematopoietic stem cells (HSCs) and in the whole BM microenvironment with an aberrant cross talk between hematopoietic elements and stromal compartment. This review highlights the role of MSCs in MDS showing functional and molecular alterations such as altered cell-cycle regulation with impaired proliferative potential, dysregulated cytokine secretion, and an abnormal gene expression profile. Here, the current knowledge of impaired functional properties of both aged MSCs and MSCs in MDS have been described with a special focus on inflammation and senescence induced changes in the BM microenvironment. Furthermore, a better understanding of aberrant BM microenvironment could improve future potential therapies.
Collapse
Affiliation(s)
- Domenico Mattiucci
- 1 Dipartimento di Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| | - Giulia Maurizi
- 1 Dipartimento di Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| | - Pietro Leoni
- 1 Dipartimento di Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| | - Antonella Poloni
- 1 Dipartimento di Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
42
|
Xiong BJ, Tan QW, Chen YJ, Zhang Y, Zhang D, Tang SL, Zhang S, Lv Q. The Effects of Platelet-Rich Plasma and Adipose-Derived Stem Cells on Neovascularization and Fat Graft Survival. Aesthetic Plast Surg 2018; 42:1-8. [PMID: 29302732 DOI: 10.1007/s00266-017-1062-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 12/13/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND Adipose-derived stem cell (ADSCs)-assisted and platelet-rich plasma (PRP)-assisted lipofilling aim to enhance angiogenesis and cell proliferation and are promising techniques for lipofilling. This study aimed to compare the outcomes of ADSCs-assisted and PRP-assisted lipofilling. METHODS Adipose tissue and human venous blood were obtained from women with early breast cancer. Human ADSCs were isolated and amplified in vitro. PRP was extracted through double centrifugation. The effect of PRP on ADSCs proliferation was evaluated. In the in vivo study, 1 ml of adipose tissue with saline (control group), PRP (PRP group), or ADSCs (ADSCs group) was injected subcutaneously into the dorsum of nude mice. At 2, 4, 8, and 12 weeks after injection, tissues were assessed for volume retention and ultrasound abnormality. For histological assessment, hematoxylin and eosin staining were performed. RESULTS Cytokines in PRP and blood were comparable. Regarding the in vitro assay, PRP significantly improved ADSCs proliferation, and the effect was dose-dependent. Concerning the in vivo study, for each time point, ADSCs-assisted lipofilling showed superior volume maintenance. Similarly, the PRP group showed improved angiogenesis and fat survival, as compared with the control group. The angiogenic effect of PRP was inferior to that of ADSCs at most time points. No significant difference was observed at 12 weeks after lipofilling. Complication rates were comparable between the PRP group and ADSCs group. CONCLUSIONS PRP-assisted and ADSCs-assisted lipofilling can significantly improve the cosmetic results of grafted fat. PRP-assisted lipofilling, which is considered convenient and clinically available, is a promising technique to improve neovascularization and fat survival. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
|
43
|
|
44
|
Mizushima T, Fukata T, Takeyama H, Takahashi H, Haraguchi N, Nishimura J, Hata T, Matsuda C, Yamamoto H, Doki Y, Mori M. The features of adipose-derived stem cells in patients with inflammatory bowel diseases. Surg Today 2017; 48:352-358. [PMID: 29027585 DOI: 10.1007/s00595-017-1591-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/13/2017] [Indexed: 12/17/2022]
Abstract
PURPOSE The treatment outcomes for inflammatory bowel disease (IBD) have been improving, but the development of better therapies is needed. Stem cell therapy is promising, but little is known about the differences in adipose-derived stem cells (ADSCs) between IBD patients and healthy individuals. METHODS ADSCs were isolated from subcutaneous adipose tissue (SAT) in IBD (Crohn's disease, 3; ulcerative colitis, 2) and non-IBD (colorectal cancer, 5; breast cancer, 1) patients. We also analyzed the effects of tumor necrosis factor (TNF)-α on murine ADSCs. RESULTS The numbers of stromal vascular fraction (SVF) cells per gram of SAT were 7.72 ± 3.03 × 105 in IBD and 8.51 ± 8.80 × 105 in non-IBD patients (p > 0.05). The proportions of ADSCs in SVF cells were 4.98 ± 2.61% in IBD and 1.02 ± 0.67% in non-IBD patients. The numbers of ADSCs per gram of SAT were 4.16 ± 2.96 × 104 in IBD and 0.88 ± 1.04 × 104 in non-IBD patients. The proportions and numbers of ADSCs were significantly higher in IBD patients than in non-IBD patients. TNF-α significantly facilitated the proliferation and motility of murine ADSCs. CONCLUSION These results showed the potential advantage of freshly isolated autologous ADSCs in IBD patients.
Collapse
Affiliation(s)
- Tsunekazu Mizushima
- Department of Therapeutics for Inflammatory Bowel Diseases, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Tadafumi Fukata
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hiroshi Takeyama
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hidekazu Takahashi
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Naotsugu Haraguchi
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Junichi Nishimura
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Taishi Hata
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Chu Matsuda
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hirofumi Yamamoto
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yuichiro Doki
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Masaki Mori
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
45
|
Liu J, Ren J, Su L, Cheng S, Zhou J, Ye X, Dong Y, Sun S, Qi F, Liu Z, Pleat J, Zhai H, Zhu N. Human adipose tissue-derived stem cells inhibit the activity of keloid fibroblasts and fibrosis in a keloid model by paracrine signaling. Burns 2017; 44:370-385. [PMID: 29029852 DOI: 10.1016/j.burns.2017.08.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 08/23/2017] [Accepted: 08/25/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Human adipose tissue-derived mesenchymal stem cells (ASCs) have potential utility as modulators of the regeneration of tissue that is inflamed or scarred secondary to injuries such as burns or trauma. However, the effect of ASCs on one particular type of scarring, keloidal disease, remains unknown. The absence of an optimal model for investigation has hindered the development of an effective therapy using ASCs for keloids. OBJECTIVE To investigate the influence of ASCs on angiogenesis, extracellular matrix deposition, and inflammatory cell influx in keloids. METHODS We analyzed the proliferation, migration, and apoptosis of human keloid-derived fibroblasts treated with a starvation-induced, conditioned medium from ASCs (ASCs-CM). This was achieved by Brdu proliferation assay, a validated co-culture migration assay, and flow cytometry, respectively. To assess the change in phenotype to a pro-fibrotic state, fibroblasts were analyzed by real-time PCR and contraction assay. A keloid implantation animal model was used to assess the paracrine effect of ASCs histochemically and immunohistochemically on scar morphology, collagen deposition, inflammatory cell composition, and blood vessel density. In tandem, an antibody-based array was used to identify protein concentration in the presence of ASCs-CM at time point 0, 24, and 48h. RESULTS ASCs-CM inhibited the proliferation and collagen synthesis of human keloid-derived fibroblasts. ASCs-CM was associated with reduced inflammation and fibrosis in the keloid implantation model. Thirty-four cytokines were differentially regulated by ASCs-CM at 24h. These included molecules associated with apoptosis, matrix metalloproteases, and their inhibitors. The same molecules were present at relatively higher concentrations at the 48h timepoint. CONCLUSION These results suggest that ASCs are associated with the inhibition of fibrosis in keloids by a paracrine effect. This phenomenon may have utility as a therapeutic approach in the clinical environment.
Collapse
Affiliation(s)
- Jianlan Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jie Ren
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lina Su
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shimeng Cheng
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jing Zhou
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaolu Ye
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yabin Dong
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Silei Sun
- Department of Emergency Intensive Care Unit, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fazhi Qi
- Department of Plastic Surgery, Zhongshan Hospital, Shanghai, China
| | - Zhifei Liu
- Plastic Surgery Department, Peking Union Medical Hospital, Chinese Academy of Medical Sciences, Beijing 100032, China
| | - Jonathon Pleat
- Department of Plastic, Reconstructive and Burns Surgery, Southmead Hospital & University of Bristol, Bristol, UK
| | - Hongjun Zhai
- Anshan Hospital of The First Hospital of China Medical University, China.
| | - Ningwen Zhu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Plastic, Reconstructive and Burns Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
46
|
Dolivo DM, Larson SA, Dominko T. Fibroblast Growth Factor 2 as an Antifibrotic: Antagonism of Myofibroblast Differentiation and Suppression of Pro-Fibrotic Gene Expression. Cytokine Growth Factor Rev 2017; 38:49-58. [PMID: 28967471 DOI: 10.1016/j.cytogfr.2017.09.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 09/22/2017] [Indexed: 02/08/2023]
Abstract
Fibrosis is a pathological condition that is characterized by the replacement of dead or damaged tissue with a nonfunctional, mechanically aberrant scar, and fibrotic pathologies account for nearly half of all deaths worldwide. The causes of fibrosis differ somewhat from tissue to tissue and pathology to pathology, but in general some of the cellular and molecular mechanisms remain constant regardless of the specific pathology in question. One of the common mechanisms underlying fibroses is the paradigm of the activated fibroblast, termed the "myofibroblast," a differentiated mesenchymal cell with demonstrated contractile activity and a high rate of collagen deposition. Fibroblast growth factor 2 (FGF2), one of the members of the mammalian fibroblast growth factor family, is a cytokine with demonstrated antifibrotic activity in non-human animal, human, and in vitro models. FGF2 is highly pleiotropic and its receptors are present on many different cell types throughout the body, lending a great deal of variety to the potential mechanisms of FGF2 effects on fibrosis. However, recent reports demonstrate that a substantial contribution to the antifibrotic effects of FGF2 comes from the inhibitory effects of FGF2 on connective tissue fibroblasts, activated myofibroblasts, and myofibroblast progenitors. FGF2 demonstrates effects antagonistic towards fibroblast activation and towards mesenchymal transition of potential myofibroblast-forming cells, as well as promotes a gene expression paradigm more reminiscent of regenerative healing, such as that which occurs in the fetal wound healing response, than fibrotic resolution. With a better understanding of the mechanisms by which FGF2 alters the wound healing cascade and results in a shift away from scar formation and towards functional tissue regeneration, we may be able to further address the critical need of therapy for varied fibrotic pathologies across myriad tissue types.
Collapse
Affiliation(s)
- David M Dolivo
- Worcester Polytechnic Institute, Department of Biology and Biotechnology,100 Institute Road, Worcester, MA, 01609, United States
| | - Sara A Larson
- Worcester Polytechnic Institute, Department of Biology and Biotechnology,100 Institute Road, Worcester, MA, 01609, United States
| | - Tanja Dominko
- Worcester Polytechnic Institute, Department of Biology and Biotechnology,100 Institute Road, Worcester, MA, 01609, United States.
| |
Collapse
|
47
|
Dedifferentiated Adipocytes Promote Adipose Tissue Generation within an External Suspension Device. Plast Reconstr Surg 2017; 140:525-536. [DOI: 10.1097/prs.0000000000003601] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
48
|
|
49
|
Wang B, Lee WYW, Huang B, Zhang JF, Wu T, Jiang X, Wang CC, Li G. Secretome of Human Fetal Mesenchymal Stem Cell Ameliorates Replicative Senescen. Stem Cells Dev 2016; 25:1755-1766. [PMID: 27539404 DOI: 10.1089/scd.2016.0079] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Autologous mesenchymal stem cells (MSC) are widely used cell source for cell-based tissue repair and regeneration, but replicative senescence and the associated loss of cellular activity during in vitro expansion limit their therapeutic potential. How to preserve or even enhance the proliferation and differentiation ability of MSC from aged donors without genetic modification remains a challenge to meet clinical need. MSC isolated from human fetal tissues (hFMSC) exhibit higher proliferation and differentiation activities even in prolonged in vitro culture, which might be modulated by autocrine/paracrine action. In the present study, we hypothesized that the bioactive factors secreted by hFMSC, collectively named as hFMSC secretome (HFS), could possess beneficial effect on human adult MSC (hAMSC) undergoing replicative senescence, thus promoting their capability of proliferation and differentiation. HFS was prepared by centrifugation of hFMSC conditioned medium, followed by column-based concentration, and the total protein content of the HFS was quantified to standardize treatment concentration. When compared with hAMSC secretome (HAS), HFS treatment significantly reduced senescence associated-β-galactosidase expression and activity (senescence marker) and enhanced cell proliferation and osteogenic differentiation potential of hAMSC in prolonged in vitro culture. Cellular studies revealed concomitant activation of sirt1 and foxo3a in hAMSC after HFS treatment, which was associated with upregulation of p21 and downregulation of bax and p53. The changes of these senescence associated markers suggested that HFS, but not HAS, could ameliorate replicative senescence of hAMSC in vitro. In nude mice, HFS pretreatment restored the osteogenic ability of senescent hAMSC. Tumor xenograft model revealed that HFS did not promote tumor growth. In conclusion, this study suggests that HFS could be an effective and safe method to overcome replicative senescence and facilitate the therapeutic potential of hAMSC.
Collapse
Affiliation(s)
- Bin Wang
- 1 Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong , Hong Kong SAR, People's Republic of China .,2 Shenzhen Research Institute, The Chinese University of Hong Kong , Shenzhen, People's Republic of China
| | - Wayne Yuk-Wai Lee
- 1 Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong , Hong Kong SAR, People's Republic of China .,2 Shenzhen Research Institute, The Chinese University of Hong Kong , Shenzhen, People's Republic of China
| | - Biao Huang
- 3 Faculty of Medicine, Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong , Hong Kong SAR, People's Republic of China .,4 Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, The Chinese University of Hong Kong , Hong Kong SAR, People's Republic of China
| | - Jin-Fang Zhang
- 1 Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong , Hong Kong SAR, People's Republic of China .,2 Shenzhen Research Institute, The Chinese University of Hong Kong , Shenzhen, People's Republic of China
| | - TianYi Wu
- 1 Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong , Hong Kong SAR, People's Republic of China .,2 Shenzhen Research Institute, The Chinese University of Hong Kong , Shenzhen, People's Republic of China
| | - Xiaohua Jiang
- 3 Faculty of Medicine, Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong , Hong Kong SAR, People's Republic of China .,4 Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, The Chinese University of Hong Kong , Hong Kong SAR, People's Republic of China
| | - Chi Chiu Wang
- 5 Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong , Hong Kong SAR, People's Republic of China .,6 Reproduction and Development, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China .,7 School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Gang Li
- 1 Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong , Hong Kong SAR, People's Republic of China .,2 Shenzhen Research Institute, The Chinese University of Hong Kong , Shenzhen, People's Republic of China .,4 Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, The Chinese University of Hong Kong , Hong Kong SAR, People's Republic of China
| |
Collapse
|
50
|
Jeong W, Yang X, Lee J, Ryoo Y, Kim J, Oh Y, Kwon S, Liu D, Son D. Serial changes in the proliferation and differentiation of adipose-derived stem cells after ionizing radiation. Stem Cell Res Ther 2016; 7:117. [PMID: 27530249 PMCID: PMC4988041 DOI: 10.1186/s13287-016-0378-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/23/2016] [Accepted: 07/27/2016] [Indexed: 02/06/2023] Open
Abstract
Background Adipose-derived stem cells (ASCs) are important to homeostasis and the regeneration of subcutaneous fat. Hence, we examined the proliferation and differentiation capacity of irradiated ASCs over time. Methods Two female pigs received a single 18 Gy dose of ionizing radiation to an 18 × 8 cm area on the dorsal body skin via a 6 MeV electron beam. After irradiation, the ASCs were cultured from adipose tissue harvested from a non-irradiated area and an irradiated area at 2, 4, and 6 weeks. The proliferation capacity of ASCs was evaluated by a colony-forming units–fibroblasts (CFUs-Fs) assay, a cholecystokinin (CCK) test with 10 % fetal bovine serum (FBS), and a 1 % FBS culture test. The senescence of ASCs was evaluated through morphological examination, immunophenotyping, and β-galactosidase activity, and the multipotent differentiation potential of ASCs was evaluated in adipogenic, osteogenic, and chondrogenic differentiation media. Results Irradiated ASCs demonstrated significantly decreased proliferative capacity 6 weeks after irradiation. As well, the cells underwent senescence, which was confirmed by blunted morphology, weak mesenchymal cell surface marker expression, and elevated β-galactosidase activity. Irradiated ASCs also exhibited significant losses in the capacity for adipocyte and chondrocyte differentiation. In contrast, osteogenic differentiation was preserved in irradiated ASCs. Conclusions We observed decreased proliferation and senescence of irradiated ASCs compared to non-irradiated ASCs 6 weeks after irradiation. Furthermore, irradiated ASCs demonstrated impaired adipocyte and chondrocyte differentiation but retained their osteogenic differentiation capacity. Our results could shed light on additional pathogenic effects of late irradiation, including subcutaneous fibrosis and calcinosis.
Collapse
Affiliation(s)
- Woonhyeok Jeong
- Department of Plastic and Reconstructive Surgery, Institute for Medical Science, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Xiao Yang
- Department of Plastic and Reconstructive Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jeongmi Lee
- Department of Plastic and Reconstructive Surgery, Institute for Medical Science, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Youngwook Ryoo
- Department of Dermatology, Institute for Medical Science, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Jinhee Kim
- Department of Radiation Oncology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Youngkee Oh
- Department of Radiation Oncology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Sunyoung Kwon
- Department of Pathology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Dalie Liu
- Department of Plastic and Reconstructive Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Daegu Son
- Department of Plastic and Reconstructive Surgery, Institute for Medical Science, Keimyung University School of Medicine, Daegu, Republic of Korea.
| |
Collapse
|