1
|
Ragni E, Taiana M, Visconte C, Kon E, Landoni S, Colombo C, Mangiavini L, Peretti G, de Girolamo L. Side-to-side characterisation of cellular content, soluble factors and in vitro potential on chondrocytes for bone marrow aspirate concentrate and adipose-derived stromal vascular fraction. J Exp Orthop 2025; 12:e70254. [PMID: 40357029 PMCID: PMC12066993 DOI: 10.1002/jeo2.70254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 05/15/2025] Open
Abstract
Purpose Orthobiologics gained popularity for the treatment of musculoskeletal pathologies, including osteoarthritis (OA). Bone marrow aspirate concentrate (BMAC) and adipose-derived stromal vascular fraction (SVF) were reported to reduce OA symptoms, contributing to the restoration of joint homoeostasis. Variability in production protocols and lack of extensive characterisation hinder a clear indication for the choice of a product over the other. The purpose of this study was to characterise side-by-side BMAC and SVF obtained with the same family of devices, by assessing cell immunophenotype, release of soluble factors and their ability to reduce inflammation in a pathologic in vitro chondrocyte model. Methods BMAC (iliac crest) and SVF (abdomen liposuction) were obtained from 28 (55 years old ± 8) and 39 patients (56 years old ± 9), with Hy-Tissue BMAC and Hy-Tissue SVF. BMAC/SVF were characterised for cellular content. The number of mesenchymal stromal cells (MSCs) was investigated by flow cytometry (CD45-CD31-CD34+CD90+CD105-/LCD146-, adipose-MSCs; CD45-CD271+, bone marrow-MSCs). Two-hundred factors (cytokines, chemokines, receptors, growth factors and inflammatory molecules) were tested by enzyme-linked immunosorbent assay (ELISA). Anti-inflammatory potential was assayed in vitro on interleukin-1 beta (IL1β)-treated chondrocytes by quantitative reverse transcription polymerase chain reaction (qRT-PCR) arrays of 84 genes involved in inflammatory processes. Results BMAC had higher concentration for white cells (213x), erythrocytes (49x) and platelets (25x), while the number of MSCs resulted comparable between the two products (1000 cells/mL). One-hundred and twenty-one soluble factors were identified in all analysed samples, with 88 more abundant in BMAC and one in SVF. Gene ontology revealed that the higher concentrated molecules were mainly growth factors and/or involved in differentiation processes. Both orthobiologics reduced inflammation in the in vitro chondrocyte model, with BMAC showing higher efficacy. Conclusions Using specific commercial systems, both orthobiologics showed anti-inflammatory effects in vitro. BMAC had higher blood cell and growth factor concentrations than SVF, with greater efficacy. However, variability in commercial systems limits generalisation, requiring further study to draw conclusions when different devices are employed. Level of Evidence NA.
Collapse
Affiliation(s)
- Enrico Ragni
- IRCCS Ospedale Galeazzi—Sant'Ambrogio, Laboratorio di Biotecnologie Applicate all'OrtopediaMilanoItaly
| | - Michela Taiana
- IRCCS Ospedale Galeazzi—Sant'Ambrogio, Laboratorio di Biotecnologie Applicate all'OrtopediaMilanoItaly
| | - Caterina Visconte
- IRCCS Ospedale Galeazzi—Sant'Ambrogio, Laboratorio di Biotecnologie Applicate all'OrtopediaMilanoItaly
| | - Elizaveta Kon
- IRCCS Humanitas Research HospitalRozzanoItaly
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
| | - Simona Landoni
- IRCCS Ospedale Galeazzi—Sant'Ambrogio, Laboratorio di Biotecnologie Applicate all'OrtopediaMilanoItaly
| | - Cecilia Colombo
- IRCCS Ospedale Galeazzi—Sant'Ambrogio, Laboratorio di Biotecnologie Applicate all'OrtopediaMilanoItaly
| | - Laura Mangiavini
- Department of Biomedical Sciences for HealthUniversity of MilanMilanItaly
- IRCCS Ospedale Galeazzi—Sant'Ambrogio, Unità Operativa E.U.O.R.R.MilanItaly
| | - Giuseppe Peretti
- Department of Biomedical Sciences for HealthUniversity of MilanMilanItaly
- IRCCS Ospedale Galeazzi—Sant'Ambrogio, Unità Operativa E.U.O.R.R.MilanItaly
| | - Laura de Girolamo
- IRCCS Ospedale Galeazzi—Sant'Ambrogio, Laboratorio di Biotecnologie Applicate all'OrtopediaMilanoItaly
| |
Collapse
|
2
|
Sueters J, van Heiningen R, de Vries R, Guler Z, Huirne J, Smit T. Advances in tissue engineering of peripheral nerve and tissue innervation - a systematic review. J Tissue Eng 2025; 16:20417314251316918. [PMID: 39911939 PMCID: PMC11795627 DOI: 10.1177/20417314251316918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
Although various options are available to treat injured organs and peripheral nerves, none is without limitations. Auto- and allografts are the first choice of treatment, but tissue survival or functionality is not guaranteed due to often limited vascular and neural networks. In response, tissue-engineered solutions have been developed, yet clinical translations is rare. In this study, a systematic review was performed on tissue-engineered advancements for peripheral nerves and tissues, to aid future developments in bridging the gap toward the clinic by identifying high-potential solutions and unexplored areas. A systematic search was performed in PubMed, Embase, Web of Science, and Scopus until November 9, 2023. Search terms involved "tissue engineering," "guided," "tissue scaffold," and "tissue graft," together with "innervation" and "reinnervation." Original in vivo or in vitro studies meeting the inclusion criteria (tissue-engineered peripheral nerve/innervation of tissue) and no exclusion criteria (no full text available; written in foreign language; nonoriginal article; tissue-engineering of central nervous system; publication before 2012; insufficient study quality or reproducibility) were assessed. A total of 68 out of 3626 original studies were included. Data extraction was based on disease model, cell origin and host species, biomaterial nature and composition, and external stimuli of biological, chemical or physical origin. Although tissue engineering is still in its infancy, explored innervation strategies of today were highlighted with respect to biomaterials, cell types, and external stimuli. The findings emphasize that natural biomaterials, pre-seeding with autologous cell sources, and solutions for reproductive organs are beneficial for future research. Natural biomaterials possess important cues required for cell-material interaction and closely resemble native tissue in terms of biomechanical, geometrical and chemical composition. Autologous cells induce biomaterial functionalization. As these solutions pose no risk of immunorejection and have demonstrated good outcomes, they are most likely to fulfill the clinical demands.
Collapse
Affiliation(s)
- Jayson Sueters
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
| | - Rowan van Heiningen
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
- Angiogenesis Laboratory, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
| | - Ralph de Vries
- Medical Library, Vrije Universiteit, Amsterdam, The Netherlands
| | - Zeliha Guler
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Obstetrics and Gynecology, Amsterdam UMC – location AMC, Amsterdam, The Netherlands
| | - Judith Huirne
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
| | - Theo Smit
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Medical Biology, Amsterdam UMC – location AMC, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Atta H, Kassem DH, Kamal MM, Hamdy NM. Harnessing the ubiquitin proteasome system as a key player in stem cell biology. Biofactors 2025; 51:e2157. [PMID: 39843166 DOI: 10.1002/biof.2157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/20/2024] [Indexed: 01/24/2025]
Abstract
Intracellular proteins take part in almost every body function; thus, protein homeostasis is of utmost importance. The ubiquitin proteasome system (UPS) has a fundamental role in protein homeostasis. Its main role is to selectively eradicate impaired or misfolded proteins, thus halting any damage that could arise from the accumulation of these malfunctioning proteins. Proteasomes have a critical role in controlling protein homeostasis in all cell types, including stem cells. We will discuss the role of UPS enzymes as well as the 26S proteasome complex in stem cell biology from several angles. First, we shall overview common trends of proteasomal activity and gene expression of different proteasomal subunits and UPS enzymes upon passaging and differentiation of stem cells toward various cell lineages. Second, we shall explore the effect of modulating proteasomal activity in stem cells and navigate through the interrelation between proteasomes' activity and various proteasome-related transcription factors. Third, we will shed light on curated microRNAs and long non-coding RNAs using various bioinformatics tools that might have a possible role in regulating UPS in stem cells and possibly, upon manipulation, can enhance the differentiation process into different lineages and/or delay senescence upon cell passaging. This will help to decipher the role played by individual UPS enzymes and subunits as well as various interrelated molecular mediators in stem cells' maintenance and/or differentiation and open new avenues in stem cell research. This can ultimately provide a leap toward developing novel therapeutic interventions related to proteasome dysregulation.
Collapse
Affiliation(s)
- Hind Atta
- School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, Cairo, Egypt
| | - Dina H Kassem
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed M Kamal
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
- Drug Research and Development Group, Health Research Center of Excellence, The British University in Egypt, Cairo, Egypt
| | - Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
4
|
Hemati S, Ghiasi M, Salimi A. Osteogenic Differentiation of Adipose Tissue-Derived Mesenchymal Stem Cells on Composite Polymeric Scaffolds: A Review. Curr Stem Cell Res Ther 2025; 20:33-49. [PMID: 38315659 DOI: 10.2174/011574888x263333231218065453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 02/07/2024]
Abstract
The mesenchymal stem cells (MSCs) are the fundamental part of bone tissue engineering for the emergence of reconstructive medicine. Bone tissue engineering has recently been considered a promising strategy for treating bone diseases and disorders. The technique needs a scaffold to provide an environment for cell attachment to maintain cell function and a rich source of stem cells combined with appropriate growth factors. MSCs can be isolated from adipose tissue (ASCs), bone marrow (BM-MSCs), or umbilical cord (UC-MSCs). In the present study, the potential of ASCs to stimulate bone formation in composite polymeric scaffolds was discussed and it showed that ASCs have osteogenic ability in vitro. The results also indicated that the ASCs have the potential for rapid growth, easier adipose tissue harvesting with fewer donor site complications and high proliferative capacity. The osteogenic differentiation capacity of ASCs varies due to the culture medium and the addition of factors that can change signaling pathways to increase bone differentiation. Furthermore, gene expression analysis has a significant impact on improving our understanding of the molecular pathways involved in ASCs and, thus, osteogenic differentiation. Adding some drugs, such as dexamethasone, to the biomaterial composite also increases the formation of osteocytes. Combining ASCs with scaffolds synthesized from natural and synthetic polymers seems to be an effective strategy for bone regeneration. Applying exopolysaccharides, such as schizophyllan, chitosan, gelatin, and alginate in composite scaffolds enhances the osteogenesis potential of ASCs in bone tissue regeneration.
Collapse
Affiliation(s)
- Saideh Hemati
- Department of Cellular and Molecular Biology, Faculty of Biology, Science and Research Branch of Islamic Azad University, Tehran, Iran
| | - Mohsen Ghiasi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Salimi
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Shen YJ, Huang YC, Cheng YC. Advancements in Antioxidant-Based Therapeutics for Spinal Cord Injury: A Critical Review of Strategies and Combination Approaches. Antioxidants (Basel) 2024; 14:17. [PMID: 39857350 PMCID: PMC11763222 DOI: 10.3390/antiox14010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/21/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Spinal cord injury (SCI) initiates a cascade of secondary damage driven by oxidative stress, characterized by the excessive production of reactive oxygen species and other reactive molecules, which exacerbate cellular and tissue damage through the activation of deleterious signaling pathways. This review provides a comprehensive and critical evaluation of recent advancements in antioxidant-based therapeutic strategies for SCI, including natural compounds, RNA-based therapies, stem cell interventions, and biomaterial applications. It emphasizes the limitations of single-regimen approaches, particularly their limited efficacy and suboptimal delivery to injured spinal cord tissue, while highlighting the synergistic potential of combination therapies that integrate multiple modalities to address the multifaceted pathophysiology of SCI. By analyzing emerging trends and current limitations, this review identifies key challenges and proposes future directions, including the refinement of antioxidant delivery systems, the development of multi-targeted approaches, and strategies to overcome the structural complexities of the spinal cord. This work underscores the pressing need for innovative and integrative therapeutic approaches to advance the clinical translation of antioxidant-based interventions and improve outcomes for SCI patients.
Collapse
Affiliation(s)
- Yang-Jin Shen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yin-Cheng Huang
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333423, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou Medical Center, Taoyuan 333423, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yi-Chuan Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333423, Taiwan
| |
Collapse
|
6
|
Ouji-Sageshima N, Hiyama A, Kumamoto M, Kitabatake M, Hara A, Furukawa R, Hontsu S, Kawaguchi T, Sawabata N, Muro S, Ito T. Adipose-Derived Mesenchymal Stem Cells (ADSCs) Have Anti-Fibrotic Effects on Lung Fibroblasts from Idiopathic Pulmonary Fibrosis (IPF) Patients. Cells 2024; 13:2050. [PMID: 39768142 PMCID: PMC11674916 DOI: 10.3390/cells13242050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/27/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common type of fibrosis in lungs, characterized as a chronic and progressive interstitial lung disease involving pathological findings of fibrosis with a median survival of 3 years. Despite the knowledge accumulated regarding IPF from basic and clinical research, an effective medical therapy for the condition remains to be established. Thus, it is necessary for further research, including stem cell therapy, which will provide new insights into and expectations for IPF treatment. Recently, it has been reported that one of the new therapeutic candidates for IPF is adipose-derived mesenchymal stem cells (ADSCs), which have several benefits, such as easy accessibility and minimal morbidity compared to bone marrow-derived mesenchymal stem cells. Therefore, we investigated the possibility of ADSCs as a therapeutic candidate for IPF. Using human lung fibroblasts (LFs) from IPF patients, we demonstrated that human IPF LFs cocultured with ADSCs led to reduced fibrosis-related genes. Further analysis revealed that ADSCs prevented the activation of the ERK signaling pathway in IPF LFs via the upregulation of protein tyrosine phosphatase receptor-type R (PTPRR), which negatively regulates the ERK signaling pathway. Moreover, we demonstrated that intravascular administration of ADSCs improved the pathogenesis of bleomycin-induced pulmonary fibrosis with reduced collagen deposition in histology and hydroxyproline quantification and collagen markers such as the gene expression of types I and III collagen and α-smooth muscle actin (α-SMA) in a murine model. ADSC transfer was also investigated in a humanized mouse model of lung fibrosis induced via the infusion of human IPF LFs, because the bleomycin installation model does not fully recapitulate the pathogenesis of IPF. Using the humanized mouse model, we found that intravascular administration of ADSCs also improved fibrotic changes in the lungs. These findings suggest that ADSCs are a promising therapeutic candidate for IPF.
Collapse
Affiliation(s)
- Noriko Ouji-Sageshima
- Department of Immunology, Nara Medical University, Kashihara 634-8521, Nara, Japan; (N.O.-S.); (A.H.); (M.K.); (A.H.); (R.F.)
| | - Aiko Hiyama
- Department of Immunology, Nara Medical University, Kashihara 634-8521, Nara, Japan; (N.O.-S.); (A.H.); (M.K.); (A.H.); (R.F.)
| | - Makiko Kumamoto
- Department of Respiratory Medicine, Nara Medical University, Kashihara 634-8521, Nara, Japan; (M.K.); (S.H.); (S.M.)
| | - Masahiro Kitabatake
- Department of Immunology, Nara Medical University, Kashihara 634-8521, Nara, Japan; (N.O.-S.); (A.H.); (M.K.); (A.H.); (R.F.)
| | - Atsushi Hara
- Department of Immunology, Nara Medical University, Kashihara 634-8521, Nara, Japan; (N.O.-S.); (A.H.); (M.K.); (A.H.); (R.F.)
| | - Ryutaro Furukawa
- Department of Immunology, Nara Medical University, Kashihara 634-8521, Nara, Japan; (N.O.-S.); (A.H.); (M.K.); (A.H.); (R.F.)
| | - Shigeto Hontsu
- Department of Respiratory Medicine, Nara Medical University, Kashihara 634-8521, Nara, Japan; (M.K.); (S.H.); (S.M.)
| | - Takeshi Kawaguchi
- Department of Thoracic and Cardiovascular Surgery, Nara Medical University, Kashihara 634-8521, Nara, Japan; (T.K.); (N.S.)
| | - Noriyoshi Sawabata
- Department of Thoracic and Cardiovascular Surgery, Nara Medical University, Kashihara 634-8521, Nara, Japan; (T.K.); (N.S.)
| | - Shigeo Muro
- Department of Respiratory Medicine, Nara Medical University, Kashihara 634-8521, Nara, Japan; (M.K.); (S.H.); (S.M.)
| | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Kashihara 634-8521, Nara, Japan; (N.O.-S.); (A.H.); (M.K.); (A.H.); (R.F.)
| |
Collapse
|
7
|
Dao HH, Nguyen TH, Hoang DH, Vu BD, Tran MA, Le MT, Hoang NTM, Bui AV, Than UTT, Nguyen XH. Manufacturing exosomes for wound healing: Comparative analysis of culture media. PLoS One 2024; 19:e0313697. [PMID: 39541412 PMCID: PMC11563385 DOI: 10.1371/journal.pone.0313697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Mesenchymal stem cell (MSC)-derived exosomes (EXs) have emerged as promising therapeutic agents for wound healing. However, the optimal conditions for manufacturing MSC-derived EXs that maximize their wound-healing potential have yet to be established. Hence, we compared the efficacy of five different MSC culture media, including three different serum-free, a platelet-supplemented, and a fetal bovine serum-supplemented media, in exosome manufacturing for wound healing applications. Although umbilical cord-derived MSCs (UCMSCs) cultured in these media exhibited similar proliferation, morphology, MSC surface marker expression, and stemness, EXs derived from UCMSCs cultured in different culture media displayed varying levels of growth factors and cytokines. Notably, EXs derived from platelet-supplemented media (DM-PLT_EXs) exhibited significantly higher concentrations of keratinocyte growth factor (KGF), vascular endothelial growth factor (VEGF-A), platelet-derived growth factor (PDGF-BB), interleukin 6 (IL-6), interleukin 7 (IL-7), and interleukin 8 (IL-8) than EXs from other media. These differences correlated with the superior capability of DM-PLT_EXs to promote human skin fibroblast proliferation and stimulate angiogenesis of human umbilical vein endothelial cells, making them a more suitable choice for wound healing applications. Our findings emphasize the significance of the culture medium selection in tailoring the therapeutic potential of UCMSC-derived EXs for wound healing.
Collapse
Affiliation(s)
- Huy Hoang Dao
- Vinmec Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | | | | | - Bach Duong Vu
- Vinmec Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Minh-Anh Tran
- Vinmec Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Mai Thi Le
- Vinmec Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
- Faculty of Biology, VNU University of Science, Vietnam National University, Hanoi, Vietnam
| | - Nhung Thi My Hoang
- Faculty of Biology, VNU University of Science, Vietnam National University, Hanoi, Vietnam
| | - Anh Viet Bui
- Vinmec Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Uyen Thi Trang Than
- Vinmec Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
- Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Xuan-Hung Nguyen
- Vinmec Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
- Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, Vietnam
- College of Health Sciences, VinUniversity, Hanoi, Vietnam
| |
Collapse
|
8
|
Silva Couto P, Stibbs DJ, Rotondi MC, Khalife R, Wolf D, Takeuchi Y, Rafiq QA. Biological differences between adult and perinatal human mesenchymal stromal cells and their impact on the manufacturing processes. Cytotherapy 2024; 26:1429-1441. [PMID: 38970611 DOI: 10.1016/j.jcyt.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 07/08/2024]
Abstract
The biological properties of human mesenchymal stromal cells (hMSCs) have been explored in over a thousand clinical trials in the last decade. Although hMSCs can be isolated from multiple sources, the degree of biological similarity between cell populations from these sources remains to be determined. A comparative study was performed investigating the growth kinetics and functionality of hMSCs isolated from adipose tissue (AT), bone marrow (BM) and umbilical cord tissue (UCT) expanded in monolayer over five passages. Adult hMSCs (AT, BM) had a slower proliferation ability than the UCT-hMSCs, with no apparent differences in their glucose consumption profile. BM-hMSCs produced higher concentrations of endogenous vascular endothelial growth factor (VEGF) compared to AT- and UCT-hMSCs. This study also revealed that UCT-hMSCs were more efficiently transduced by a lentiviral vector carrying a VEGF gene than their adult counterparts. Following cellular immunophenotypic characterization, no differences across the sources were found in the expression levels of the typical markers used to identify hMSCs. This work established a systematic approach for cell source selection depending on the hMSC's intended clinical application.
Collapse
Affiliation(s)
- Pedro Silva Couto
- Department of Biochemical Engineering, University College London, London, UK
| | - Dale J Stibbs
- Department of Biochemical Engineering, University College London, London, UK
| | - Marco C Rotondi
- Department of Biochemical Engineering, University College London, London, UK
| | - Rana Khalife
- Department of Biochemical Engineering, University College London, London, UK
| | | | - Yasuhiro Takeuchi
- Division of Infection and Immunity, University College London, London, UK; Biotherapeutics and Advanced Therapies, Scientific Research and Innovation, Medicines and Healthcare products Regulatory Agency, Potters Bar, UK
| | - Qasim A Rafiq
- Department of Biochemical Engineering, University College London, London, UK.
| |
Collapse
|
9
|
Chen CH, Dash BS, Ting WC, Chen JP. Bone Tissue Engineering with Adipose-Derived Stem Cells in Polycaprolactone/Graphene Oxide/Dexamethasone 3D-Printed Scaffolds. ACS Biomater Sci Eng 2024; 10:6425-6440. [PMID: 39226111 DOI: 10.1021/acsbiomaterials.4c00774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
We fabricated three-dimensional (3D)-printed polycaprolactone (PCL) and PCL/graphene oxide (GO) (PGO) scaffolds for bone tissue engineering. An anti-inflammatory and pro-osteogenesis drug dexamethasone (DEX) was adsorbed onto GO and a 3D-printed PGO/DEX (PGOD) scaffold successfully improved drug delivery with a sustained release of DEX from the scaffold up to 1 month. The physicochemical properties of the PCL, PGO, and PGOD scaffolds were characterized by various analytical techniques. The biological response of these scaffolds was studied for adherence, proliferation, and osteogenic differentiation of seeded rabbit adipose-derived stem cells (ASCs) from DNA assays, alkaline phosphatase (ALP) production, calcium quantification, osteogenic gene expression, and immunofluorescence staining of osteogenic marker proteins. The PGOD scaffold was demonstrated to be the best scaffold for maintaining cell viability, cell proliferation, and osteogenic differentiation of ASCs in vitro. In vivo biocompatibility of PGOD was confirmed from subcutaneous implantation in nude mice where ASC-seeded PGOD can form ectopic bones, demonstrated by microcomputed tomography (micro-CT) analysis and immunofluorescence staining. Furthermore, implantation of PGOD/ASCs constructs into critical-sized cranial bone defects in rabbits form tissue-engineered bones at the defect site, observed using micro-CT and histological analysis.
Collapse
Affiliation(s)
- Chih-Hao Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Taoyuan, Kwei-San 33302, Taiwan
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Keelung, Keelung 20401, Taiwan
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University School of Medicine, Taoyuan, Kwei-San 33305, Taiwan
| | - Banendu Sunder Dash
- Department of Chemical and Materials Engineering, Chang Gung University, Taoyuan, Kwei-San 33302, Taiwan
| | - Wei-Chun Ting
- Department of Chemical and Materials Engineering, Chang Gung University, Taoyuan, Kwei-San 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Taoyuan, Kwei-San 33302, Taiwan
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University School of Medicine, Taoyuan, Kwei-San 33305, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Kwei-San 33305, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33302, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
| |
Collapse
|
10
|
Liu Y, Zhao C, Zhang R, Pang Y, Li L, Feng S. Progression of mesenchymal stem cell regulation on imbalanced microenvironment after spinal cord injury. Stem Cell Res Ther 2024; 15:343. [PMID: 39354635 PMCID: PMC11446099 DOI: 10.1186/s13287-024-03914-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 09/01/2024] [Indexed: 10/03/2024] Open
Abstract
Spinal cord injury (SCI) results in significant neural damage and inhibition of axonal regeneration due to an imbalanced microenvironment. Extensive evidence supports the efficacy of mesenchymal stem cell (MSC) transplantation as a therapeutic approach for SCI. This review aims to present an overview of MSC regulation on the imbalanced microenvironment following SCI, specifically focusing on inflammation, neurotrophy and axonal regeneration. The application, limitations and future prospects of MSC transplantation are discussed as well. Generally, a comprehensive perspective is provided for the clinical translation of MSC transplantation for SCI.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
- Institute of Medical Sciences, The Second Hospital of Shandong University, Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China
| | - Chenxi Zhao
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Rong Zhang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Yilin Pang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Linquan Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Shiqing Feng
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China.
- Institute of Medical Sciences, The Second Hospital of Shandong University, Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China.
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
11
|
Zangiabadi I, Ilaghi M, Shamsara A, Eftekhar‐Vaghefi SH, Saheli M, Shabani M. Exploring the impact of acetylsalicylic acid and conditioned medium obtained from mesenchymal cells, individually and in combination, on cognitive function, histological changes, and oxidant-antioxidant balance in male rats with hippocampal injury. Brain Behav 2024; 14:e70010. [PMID: 39262160 PMCID: PMC11391022 DOI: 10.1002/brb3.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/16/2024] [Accepted: 08/03/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND The hippocampus is susceptible to damage, leading to negative impacts on cognition. Conditioned medium (CM) obtained from adipose tissue-derived mesenchymal stem cells (MSCs) and acetylsalicylic acid (ASA) have shown neuroprotective effects independently. This study explored the synergistic potential of ASA and CM from adipose-derived MSCs against hippocampal injury. METHODS Adult male Wistar rats received bilateral hippocampal ethidium bromide (EB) injections to induce hippocampal damage. Rats were treated with ASA and/or CM derived from adipose tissue MSCs every 48 h for 16 days. Behavioral tests (open field test, Morris water maze, novel object recognition, and passive avoidance), oxidative stress, Western blot analysis of brain-derived neurotrophic factor (BDNF) and cerebral dopamine neurotrophic factor (CDNF) expression, and hippocampal histological investigation were conducted. RESULTS Administration of EB caused impairments in spatial, recognition, and passive avoidance memory, as well as heightened oxidative stress, reduced BDNF/CDNF expression, and pyramidal cell loss in the hippocampal CA1 region. Administration of ASA, CM, or a combination of both mitigated these hippocampal damages and cognitive deficits, elevated BDNF and CDNF levels, and alleviated the CA1 necrosis caused by EB. Moreover, co-administering ASA and CM resulted in greater improvements in spatial memory compared to administering ASA alone, suggesting possible synergistic interactions. CONCLUSIONS The ability of ASA, CM obtained from adipose tissue-derived MSCs, and their combination therapy to alleviate hippocampal injuries highlights their promising therapeutic potential as a neuroprotection strategy against brain damage. Our findings provide preliminary evidence of the potential synergistic effects of ASA and CM, which warrants further investigations.
Collapse
Affiliation(s)
- Iman Zangiabadi
- Department of Anatomy, Afzalipour School of MedicineKerman University of Medical SciencesKermanIran
| | - Mehran Ilaghi
- Institute of Neuropharmacology, Kerman Neuroscience Research CenterKerman University of Medical SciencesKermanIran
| | - Ali Shamsara
- Department of Anatomy, Afzalipour School of MedicineKerman University of Medical SciencesKermanIran
- Institute of Neuropharmacology, Kerman Neuroscience Research CenterKerman University of Medical SciencesKermanIran
| | | | - Mona Saheli
- Department of Anatomy, Afzalipour School of MedicineKerman University of Medical SciencesKermanIran
| | - Mohammad Shabani
- Institute of Neuropharmacology, Kerman Neuroscience Research CenterKerman University of Medical SciencesKermanIran
| |
Collapse
|
12
|
Ahmad N, Anker A, Klein S, Dean J, Knoedler L, Remy K, Pagani A, Kempa S, Terhaag A, Prantl L. Autologous Fat Grafting-A Panacea for Scar Tissue Therapy? Cells 2024; 13:1384. [PMID: 39195271 PMCID: PMC11352477 DOI: 10.3390/cells13161384] [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: 06/09/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024] Open
Abstract
Scars may represent more than a cosmetic concern for patients; they may impose functional limitations and are frequently associated with the sensation of itching or pain, thus impacting both psychological and physical well-being. From an aesthetic perspective, scars display variances in color, thickness, texture, contour, and their homogeneity, while the functional aspect encompasses considerations of functionality, pliability, and sensory perception. Scars located in critical anatomic areas have the potential to induce profound impairments, including contracture-related mobility restrictions, thereby significantly impacting daily functioning and the quality of life. Conventional approaches to scar management may suffice to a certain extent, yet there are cases where tailored interventions are warranted. Autologous fat grafting emerges as a promising therapeutic avenue in such instances. Fundamental mechanisms underlying scar formation include chronic inflammation, fibrogenesis and dysregulated wound healing, among other contributing factors. These mechanisms can potentially be alleviated through the application of adipose-derived stem cells, which represent the principal cellular component utilized in the process of lipofilling. Adipose-derived stem cells possess the capacity to secrete proangiogenic factors such as fibroblast growth factor, vascular endothelial growth factor and hepatocyte growth factor, as well as neurotrophic factors, such as brain-derived neurotrophic factors. Moreover, they exhibit multipotency, remodel the extracellular matrix, act in a paracrine manner, and exert immunomodulatory effects through cytokine secretion. These molecular processes contribute to neoangiogenesis, the alleviation of chronic inflammation, and the promotion of a conducive milieu for wound healing. Beyond the obvious benefit in restoring volume, the adipose-derived stem cells and their regenerative capacities facilitate a reduction in pain, pruritus, and fibrosis. This review elucidates the regenerative potential of autologous fat grafting and its beneficial and promising effects on both functional and aesthetic outcomes when applied to scar tissue.
Collapse
Affiliation(s)
- Nura Ahmad
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| | - Alexandra Anker
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| | - Silvan Klein
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| | - Jillian Dean
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Leonard Knoedler
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| | - Katya Remy
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Andrea Pagani
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| | - Sally Kempa
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| | - Amraj Terhaag
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| | - Lukas Prantl
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| |
Collapse
|
13
|
Hisamatsu D, Ikeba A, Yamato T, Mabuchi Y, Watanabe M, Akazawa C. Optimization of transplantation methods using isolated mesenchymal stem/stromal cells: clinical trials of inflammatory bowel diseases as an example. Inflamm Regen 2024; 44:37. [PMID: 39152520 PMCID: PMC11328379 DOI: 10.1186/s41232-024-00350-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/26/2024] [Indexed: 08/19/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are distributed in various tissues and are used in clinical applications as a source of transplanted cells because of their easy harvestability. Although MSCs express numerous cell-surface antigens, single-cell analyses have revealed a highly heterogeneous cell population depending on the original tissue and donor conditions, including age and interindividual differences. This heterogeneity leads to differences in their functions, such as multipotency and immunomodulatory effects, making it challenging to effectively treat targeted diseases. The therapeutic efficacy of MSCs is controversial and depends on the implantation site. Thus, there is no established recipe for the transplantation of MSCs (including the type of disease, type of origin, method of cell culture, form of transplanted cells, and site of delivery). Our recent preclinical study identified appropriate MSCs and their suitable transplantation routes in a mouse model of inflammatory bowel disease (IBD). Three-dimensional (3D) cultures of MSCs have been demonstrated to enhance their properties and sustain engraftment at the lesion site. In this note, we explore the methods of MSC transplantation for treating IBDs, especially Crohn's disease, from clinical trials published over the past decade. Given the functional changes in MSCs in 3D culture, we also investigate the clinical trials using 3D constructs of MSCs and explore suitable diseases that might benefit from this approach. Furthermore, we discuss the advantages of the prospective isolation of MSCs in terms of interindividual variability. This note highlights the need to define the method of MSC transplantation, including interindividual variability, the culture period, and the transplantation route.
Collapse
Affiliation(s)
- Daisuke Hisamatsu
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Akimi Ikeba
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Taku Yamato
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Yo Mabuchi
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, Tokyo, Japan
| | - Mamoru Watanabe
- Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
| | - Chihiro Akazawa
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
14
|
Schumacher A, Mucha P, Puchalska I, Deptuła M, Wardowska A, Tymińska A, Filipowicz N, Mieczkowska A, Sachadyn P, Piotrowski A, Pikuła M, Cichorek M. Angiopoietin-like growth factor-derived peptides as biological activators of adipose-derived mesenchymal stromal cells. Biomed Pharmacother 2024; 177:117052. [PMID: 38943988 DOI: 10.1016/j.biopha.2024.117052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024] Open
Abstract
Adipose-derived mesenchymal stromal cells (AD-MSCs) are an essential issue in modern medicine. Extensive preclinical and clinical studies have shown that mesenchymal stromal/stem cells, including AD-MSCs, have specific properties (ability to differentiate into other cells, recruitment to the site of injury) of particular importance in the regenerative process. Ongoing research aims to elucidate factors supporting AD-MSC culture and differentiation in vitro. Angiopoietin-like proteins (ANGPTLs), known for their pleiotropic effects in lipid and glucose metabolism, may play a significant role in this context. Regeneration is a complex and dynamic process controlled by many factors. ANGPTL6 (Angiopoietin-related growth factor, AGF), among many activities modulated the biological activity of stem cells. This study examined the influence of synthesized AGF-derived peptides, designated as AGF9 and AGF27, on AD-MSCs. AGF9 and AGF27 enhanced the viability and migration of AD-MSCs and acted as a chemotactic factor for these cells. AGF9 stimulated chondrogenesis and lipid synthesis during AD-MSCs differentiation, influenced AD-MSCs cytokine secretion and modulated transcriptome for such basic cell activities as migration, transport of molecules, and apoptosis. The ability of AGF9 to modulate the biological activity of AD-MSCs warrants the consideration of this peptide a noteworthy therapeutic agent that deserves further investigation for applications in regenerative medicine.
Collapse
Affiliation(s)
- Adriana Schumacher
- Division of Embryology, Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland
| | - Piotr Mucha
- Department of Molecular Biochemistry, University of Gdansk, Wita Stwosza 63 St, Gdansk 80-308, Poland
| | - Izabela Puchalska
- Department of Molecular Biochemistry, University of Gdansk, Wita Stwosza 63 St, Gdansk 80-308, Poland
| | - Milena Deptuła
- Division of Embryology, Laboratory of Tissue Engineering and Regenerative Medicine Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland
| | - Anna Wardowska
- Department of Physiopathology, Medical University of Gdansk, Debinki 7 St, Gdansk 80-211, Poland
| | - Agata Tymińska
- Division of Embryology, Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland
| | - Natalia Filipowicz
- International Research Agenda 3P- Medicine Laboratory, Medical University of Gdansk, Debinki 7 St, Gdansk 80-211, Poland
| | - Alina Mieczkowska
- International Research Agenda 3P- Medicine Laboratory, Medical University of Gdansk, Debinki 7 St, Gdansk 80-211, Poland
| | - Paweł Sachadyn
- Laboratory for Regenerative Biotechnology, Gdansk University of Technology, Narutowicza 11/12 St, Gdansk 80-233, Poland
| | - Arkadiusz Piotrowski
- International Research Agenda 3P- Medicine Laboratory, Medical University of Gdansk, Debinki 7 St, Gdansk 80-211, Poland
| | - Michał Pikuła
- Division of Embryology, Laboratory of Tissue Engineering and Regenerative Medicine Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland
| | - Miroslawa Cichorek
- Division of Embryology, Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland.
| |
Collapse
|
15
|
Gallardo J, Berríos-Cárcamo P, Ezquer F. Mesenchymal stem cells as a promising therapy for alcohol use disorder. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 178:179-211. [PMID: 39523054 DOI: 10.1016/bs.irn.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Alcohol Use Disorder (AUD) is a highly prevalent medical condition characterized by impaired control over alcohol consumption, despite negative consequences on the individual's daily life and health. There is increasing evidence suggesting that chronic alcohol intake, like other addictive drugs, induces neuroinflammation and oxidative stress, disrupting glutamate homeostasis in the main brain areas related to drug addiction. This review explores the potential application of mesenchymal stem cells (MSCs)-based therapy for the treatment of AUD. MSCs secrete a broad array of anti-inflammatory and antioxidant molecules, thus, the administration of MSCs, or their secretome, could reduce neuroinflammation and oxidative stress in the brain. These effects correlate with an increase in the expression of the main glutamate transporter, GLT1, which, through the normalization of the extracellular glutamate levels, could mediate the inhibitory effect of MSCs' secretome on chronic alcohol consumption, thus highlighting GLT1 as a central target to reduce chronic alcohol consumption.
Collapse
Affiliation(s)
- Javiera Gallardo
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Pablo Berríos-Cárcamo
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile; Research Center for the Development of Novel Therapeutics Alternatives for Alcohol Use Disorders, Santiago, Chile.
| |
Collapse
|
16
|
Gharib OA, Fahmy HA, Abdou FY. Role of Olive Leaf Extract, Mesenchymal Stem Cells or Low Radiation Dose in Alleviating Hepatic Injury in Rats. Dose Response 2024; 22:15593258241289301. [PMID: 39483141 PMCID: PMC11526167 DOI: 10.1177/15593258241289301] [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: 06/14/2024] [Accepted: 08/28/2024] [Indexed: 11/03/2024] Open
Abstract
Objectives This study was conducted to determine the efficacy of mesenchymal stem cells (MSCs) or low-dose gamma radiation (LDR) on liver injury compared to the effect of olive leaf extract as a hepatoprotective agent. Methods Rats were allocated into six groups; group I served as the negative control. Group II received 5% dextran sodium sulfate (DSS) in its drinking water for 1 week. Group III was injected with a single dose of 1 × 106 bone marrow-derived mesenchymal stem cells (BM-MSCs) intravenously. Group IV was treated as in group III after 5% DSS treatment. Group V was given 5% DSS, followed by olive leaf extract (OLE) (1000 mg/ kg, oral). Group VI: 5% DSS for 1 week, then was exposed to low-dose gamma radiation (LDR) (0.05 Gy). Results Rats treated with OLE, BM-MSCs, or exposed to LDR exerted significant alleviation in all hepatic biomarkers, significant enhancements in oxidative stress parameters, and improvements in inflammatory biomarkers Interleukin-1 beta (IL-1β) and Interferon gamma (INF-γ) hepatic contents compared with those of the DSS group. Histological pictures emphasized the biochemical findings. Conclusions BM-MSCs might be a valuable therapeutic approach to overcome hepatic injury. Exposure to LDR provided protective mechanisms that allow the body to survive better.
Collapse
Affiliation(s)
- Ola A. Gharib
- Drug Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Hanan A. Fahmy
- Drug Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Fatma Y. Abdou
- Drug Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| |
Collapse
|
17
|
Lau CS, Park SY, Ethiraj LP, Singh P, Raj G, Quek J, Prasadh S, Choo Y, Goh BT. Role of Adipose-Derived Mesenchymal Stem Cells in Bone Regeneration. Int J Mol Sci 2024; 25:6805. [PMID: 38928517 PMCID: PMC11204188 DOI: 10.3390/ijms25126805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Bone regeneration involves multiple factors such as tissue interactions, an inflammatory response, and vessel formation. In the event of diseases, old age, lifestyle, or trauma, bone regeneration can be impaired which could result in a prolonged healing duration or requiring an external intervention for repair. Currently, bone grafts hold the golden standard for bone regeneration. However, several limitations hinder its clinical applications, e.g., donor site morbidity, an insufficient tissue volume, and uncertain post-operative outcomes. Bone tissue engineering, involving stem cells seeded onto scaffolds, has thus been a promising treatment alternative for bone regeneration. Adipose-derived mesenchymal stem cells (AD-MSCs) are known to hold therapeutic value for the treatment of various clinical conditions and have displayed feasibility and significant effectiveness due to their ease of isolation, non-invasive, abundance in quantity, and osteogenic capacity. Notably, in vitro studies showed AD-MSCs holding a high proliferation capacity, multi-differentiation potential through the release of a variety of factors, and extracellular vesicles, allowing them to repair damaged tissues. In vivo and clinical studies showed AD-MSCs favoring better vascularization and the integration of the scaffolds, while the presence of scaffolds has enhanced the osteogenesis potential of AD-MSCs, thus yielding optimal bone formation outcomes. Effective bone regeneration requires the interplay of both AD-MSCs and scaffolds (material, pore size) to improve the osteogenic and vasculogenic capacity. This review presents the advances and applications of AD-MSCs for bone regeneration and bone tissue engineering, focusing on the in vitro, in vivo, and clinical studies involving AD-MSCs for bone tissue engineering.
Collapse
Affiliation(s)
- Chau Sang Lau
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - So Yeon Park
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Lalith Prabha Ethiraj
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Priti Singh
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Grace Raj
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Jolene Quek
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (Y.C.)
| | - Somasundaram Prasadh
- Center for Clean Energy Engineering, University of Connecticut, Storrs, CT 06269, USA;
| | - Yen Choo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (Y.C.)
| | - Bee Tin Goh
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
18
|
Ahmadian F, Irani M, Mohammadi-Sangcheshmeh A. Effect of exogenous genistein on osteogenic differentiation of adipose-derived mesenchymal stem cells in laying hens. Tissue Cell 2024; 87:102299. [PMID: 38228028 DOI: 10.1016/j.tice.2023.102299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/21/2023] [Accepted: 12/31/2023] [Indexed: 01/18/2024]
Abstract
Previous literature revealed that genistein might play a preventive role in osteoporosis. Therefore, we aimed to evaluate the effect of genistein on the osteogenic potency of laying hens' adipose-derived stem cells (LHASCs). The viability of LHASCs after isolation was investigated on tissue culture plastic (TCP) under exposure to genistein up to 50 μg/mL by MTT assay. Our preliminary result revealed that LHASCs cultured under genistein exposure up to 20 μg/mL are feasible. Then, we evaluated the osteogenic induction of LHASCs under exposure to 0, 10, and 20 μg/mL genistein. The Alizarin Red staining confirmed the calcium deposition. Our findings showed that osteogenic differentiation under exposure to 20 μg/mL genistein led to higher ALP activity and more calcium content. We then tried to see the probable additive effect of the genistein-plus Poly-L-lactic acid (PLLA) scaffold on the cell viability and osteogenic capacity of LHASCs. For this, cells were cultured on a PLLA scaffold and exposed to 20 μg/mL genistein. Cell growth rate, as indicated by the MTT assay, revealed no differences between the groups. LHASCs cultured on a genistein-plus PLLA scaffold showed higher ALP activity and more calcium content. The expressions of Osteocalcin, COL1A2, ALP, and Runx2 genes were increased in the genistein-plus PLLA group as compared with PLLA and TCP groups. Adequate proliferation rates and higher expression of osteogenic markers provide genistein as a suitable substrate to support the proliferation and differentiation of LHASCs. Genistein supports osteogenic induction as a further positive effect if such a PLLA scaffold is available.
Collapse
Affiliation(s)
- Farhang Ahmadian
- Department of Animal Science, Qaemshahr Branch, Islamic Azad University, Qaemshahr, Iran
| | - Mehrdad Irani
- Department of Animal Science, Qaemshahr Branch, Islamic Azad University, Qaemshahr, Iran.
| | - Abdollah Mohammadi-Sangcheshmeh
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran; Chaltasian Agri.-Animal Production Complex, Varamin, Tehran, Iran
| |
Collapse
|
19
|
Truchan K, Osyczka AM. Noggin promotes osteogenesis in human adipose-derived mesenchymal stem cells via FGFR2/Src/Akt and ERK signaling pathway. Sci Rep 2024; 14:6724. [PMID: 38509118 PMCID: PMC10954655 DOI: 10.1038/s41598-024-56858-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The balance between Noggin and bone morphogenetic proteins (BMPs) is important during early development and skeletal regenerative therapies. Noggin binds BMPs in the extracellular space, thereby preventing BMP signaling. However, Noggin may affect cell response not necessarily through the modulation of BMP signaling, raising the possibility of direct Noggin signaling through yet unspecified receptors. Here we show that in osteogenic cultures of adipose-derived stem cells (ASCs), Noggin activates fibroblast growth factor receptors (FGFRs), Src/Akt and ERK kinases, and it stabilizes TAZ proteins in the presence of dexamethasone. Overall, this leads ASCs to increased expression of osteogenic markers and robust mineral deposition. Our results also indicate that Noggin can induce osteogenic genes expression in normal human bone marrow stem cells and alkaline phosphatase activity in normal human dental pulp stem cells. Besides, Noggin can specifically activate FGFR2 in osteosarcoma cells. We believe our findings open new research avenues to further explore the involvement of Noggin in cell fate modulation by FGFR2/Src/Akt/ERK signaling and potential applications of Noggin in bone regenerative therapies.
Collapse
Affiliation(s)
- Karolina Truchan
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa St. 9, 30-387, Kraków, Poland.
| | - Anna Maria Osyczka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa St. 9, 30-387, Kraków, Poland.
| |
Collapse
|
20
|
Maličev E, Jazbec K. An Overview of Mesenchymal Stem Cell Heterogeneity and Concentration. Pharmaceuticals (Basel) 2024; 17:350. [PMID: 38543135 PMCID: PMC10975472 DOI: 10.3390/ph17030350] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/22/2024] [Accepted: 03/05/2024] [Indexed: 01/06/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are of great interest in cell therapies due to the immunomodulatory and other effects they have after autologous or allogeneic transplantation. In most clinical applications, a high number of MSCs is required; therefore, the isolated MSC population must be expanded in the cell culture until the desired number is reached. Analysing freshly isolated MSCs is challenging due to their rareness and heterogeneity, which is noticeable among donors, tissues, and cell subpopulations. Although the phenotype of MSCs in tissue can differ from those of cultured cells, phenotyping and counting are usually performed only after MSC proliferation. As MSC applicability is a developing and growing field, there is a need to implement phenotyping and counting methods for freshly isolated MSCs, especially in new one-step procedures where isolated cells are implanted immediately without cell culturing. Only by analysing harvested cells can we correctly evaluate such studies. This review describes multilevel heterogeneity and concentrations of MSCs and different strategies for phenotype determination and enumeration of freshly isolated MSCs.
Collapse
Affiliation(s)
- Elvira Maličev
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, 1000 Ljubljana, Slovenia;
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva ulica 101, 1000 Ljubljana, Slovenia
| | - Katerina Jazbec
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, 1000 Ljubljana, Slovenia;
| |
Collapse
|
21
|
Biniazan F, Stoian A, Haykal S. Adipose-Derived Stem Cells: Angiogenetic Potential and Utility in Tissue Engineering. Int J Mol Sci 2024; 25:2356. [PMID: 38397032 PMCID: PMC10889096 DOI: 10.3390/ijms25042356] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Adipose tissue (AT) is a large and important energy storage organ as well as an endocrine organ with a critical role in many processes. Additionally, AT is an enormous and easily accessible source of multipotent cell types used in our day for all types of tissue regeneration. The ability of adipose-derived stem cells (ADSCs) to differentiate into other types of cells, such as endothelial cells (ECs), vascular smooth muscle cells, or cardiomyocytes, is used in tissue engineering in order to promote/stimulate the process of angiogenesis. Being a key for future successful clinical applications, functional vascular networks in engineered tissue are targeted by numerous in vivo and ex vivo studies. The article reviews the angiogenic potential of ADSCs and explores their capacity in the field of tissue engineering (TE).
Collapse
Affiliation(s)
- Felor Biniazan
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
| | - Alina Stoian
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
| | - Siba Haykal
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada; (F.B.); (A.S.)
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Toronto, 200 Elizabeth Street Suite 8N-869, Toronto, ON M5G2C4, Canada
| |
Collapse
|
22
|
Dey MK, Devireddy RV. Adult Stem Cells Freezing Processes and Cryopreservation Protocols. Methods Mol Biol 2024; 2783:53-89. [PMID: 38478226 DOI: 10.1007/978-1-0716-3762-3_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
The development of simple but effective storage protocols for adult stem cells will greatly enhance their use and utility in tissue-engineering applications. Cryopreservation has shown the most promise but is a fairly complex process, necessitating the use of chemicals called cryoprotective agents (CPAs), freezing equipment, and obviously, storage in liquid nitrogen. The purpose of this chapter is to present a general overview of cryopreservation storage techniques and the optimal protocols/results obtained in our laboratory for long-term storage of adult stem cells using freezing storage.
Collapse
Affiliation(s)
- Mohan Kumar Dey
- Bioengineering Laboratory, Department of Mechanical Engineering, Louisiana State University, Baton Rouge, LA, USA
| | - Ram V Devireddy
- Bioengineering Laboratory, Department of Mechanical Engineering, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
23
|
Zhang W, Lee PL, Li J, Komatsu C, Wang Y, Sun H, DeSanto M, Washington K, Gorantla V, Kokai L, Solari MG. Local Delivery of Adipose Stem Cells Promotes Allograft Survival in a Rat Hind-Limb Model of Vascularized Composite Allotransplantation. Plast Reconstr Surg 2024; 153:79e-90e. [PMID: 37014960 DOI: 10.1097/prs.0000000000010510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2023]
Abstract
BACKGROUND Adipose stem cells (ASCs) are a promising cell-based immunotherapy because of their minimally invasive harvest, high yield, and immunomodulatory capacity. In this study, the authors investigated the effects of local versus systemic ASC delivery on vascularized composite allotransplant survival and alloimmune regulation. METHODS Lewis rats received hind-limb transplants from Brown Norway rats and were administered donor-derived ASCs (passage 3 or 4, 1 × 10 6 cells/rat) locally in the allograft, or contralateral limb, or systemically at postoperative day 1. Recipients were treated intraperitoneally with rabbit anti-rat lymphocyte serum on postoperative days 1 and 4 and daily tacrolimus for 21 days. Limb allografts were monitored for clinical signs of rejection. Donor cell chimerism, immune cell differentiation, and cytokine expression in recipient lymphoid organs were measured by flow cytometric analysis. The immunomodulation function of ASCs was tested by mixed lymphocyte reaction assay and ASC stimulation studies. RESULTS Local-ASC-treated recipients achieved significant prolonged allograft survival (85.7% survived >130 days; n = 6) compared with systemic-ASC and contralateral-ASC groups. Secondary donor skin allografts transplanted to the local-ASC long-term surviving recipients accepted permanently without additional immunosuppression. The increases in donor cell chimerism and regulatory T-cells were evident in blood and draining lymph nodes of the local-ASC group. Moreover, mixed lymphocyte reaction showed that ASCs inhibited donor-specific T-cell proliferation independent of direct ASC-T-cell contact. ASCs up-regulated antiinflammatory molecules in response to cytokine stimulation in vitro. CONCLUSION Local delivery of ASCs promoted long-term survival and modulated alloimmune responses in a full major histocompatibility complex-mismatched vascularized composite allotransplantation model and was more effective than systemic administration. CLINICAL RELEVANCE STATEMENT ASCs are a readily available and abundant source of therapeutic cells that could decrease the amount of systemic immunosuppression required to maintain limb and face allografts.
Collapse
Affiliation(s)
- Wensheng Zhang
- From the Department of Plastic Surgery
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
- Wilford Hall Ambulatory Surgical Center, 59th Medical Wing Office of Science and Technology, Joint Base San Antonio
| | | | - Jingjing Li
- From the Department of Plastic Surgery
- Department of Burn and Plastic Surgery, Xiangya Hospital, Central South University
| | | | - Yong Wang
- Department of Surgery, Division of Plastic and Reconstructive Surgery, University of Colorado Anschutz Medical Campus
| | | | - Marisa DeSanto
- Ohio University Heritage College of Osteopathic Medicine
| | - Kia Washington
- Department of Surgery, Division of Plastic and Reconstructive Surgery, University of Colorado Anschutz Medical Campus
| | - Vijay Gorantla
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
- Institute for Regenerative Medicine, Wake Forest School of Medicine
| | - Lauren Kokai
- From the Department of Plastic Surgery
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
| | - Mario G Solari
- From the Department of Plastic Surgery
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
| |
Collapse
|
24
|
Arderiu G, Civit-Urgell A, Badimon L. Adipose-Derived Stem Cells to Treat Ischemic Diseases: The Case of Peripheral Artery Disease. Int J Mol Sci 2023; 24:16752. [PMID: 38069074 PMCID: PMC10706341 DOI: 10.3390/ijms242316752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Critical limb ischemia incidence and prevalence have increased over the years. However, there are no successful treatments to improve quality of life and to reduce the risk of cardiovascular and limb events in these patients. Advanced regenerative therapies have focused their interest on the generation of new blood vessels to repair tissue damage through the use of stem cells. One of the most promising sources of stem cells with high potential in cell-based therapy is adipose-derived stem cells (ASCs). ASCs are adult mesenchymal stem cells that are relatively abundant and ubiquitous and are characterized by a multilineage capacity and low immunogenicity. The proangiogenic benefits of ASCs may be ascribed to: (a) paracrine secretion of proangiogenic molecules that may stimulate angiogenesis; (b) secretion of microvesicles/exosomes that are also considered as a novel therapeutic prospect for treating ischemic diseases; and (c) their differentiation capability toward endothelial cells (ECs). Although we know the proangiogenic effects of ASCs, the therapeutic efficacy of ASCs after transplantation in peripheral artery diseases patients is still relatively low. In this review, we evidence the potential therapeutic use of ASCs in ischemic regenerative medicine. We also highlight the main challenges in the differentiation of these cells into functional ECs. However, significant efforts are still needed to ascertain relevant transcription factors, intracellular signaling and interlinking pathways in endothelial differentiation.
Collapse
Affiliation(s)
- Gemma Arderiu
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau Barcelona, 08041 Barcelona, Spain; (A.C.-U.); (L.B.)
- Ciber CV, Instituto Carlos III, 28029 Madrid, Spain
| | - Anna Civit-Urgell
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau Barcelona, 08041 Barcelona, Spain; (A.C.-U.); (L.B.)
- Facultat de Medicina i Ciències de la Salut—Campus Clínic, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Lina Badimon
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau Barcelona, 08041 Barcelona, Spain; (A.C.-U.); (L.B.)
- Ciber CV, Instituto Carlos III, 28029 Madrid, Spain
| |
Collapse
|
25
|
Baron M, Drohat P, Crawford B, Hornicek FJ, Best TM, Kouroupis D. Mesenchymal Stem/Stromal Cells: Immunomodulatory and Bone Regeneration Potential after Tumor Excision in Osteosarcoma Patients. Bioengineering (Basel) 2023; 10:1187. [PMID: 37892917 PMCID: PMC10604230 DOI: 10.3390/bioengineering10101187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Osteosarcoma (OS) is a type of bone cancer that is derived from primitive mesenchymal cells typically affecting children and young adults. The current standard of treatment is a combination of neoadjuvant chemotherapy and surgical resection of the cancerous bone. Post-resection challenges in bone regeneration arise. To determine the appropriate amount of bone to be removed, preoperative imaging techniques such as bone and CT scans are employed. To prevent local recurrence, the current standard of care suggests maintaining bony and soft tissue margins from 3 to 7 cm beyond the tumor. The amount of bone removed in an OS patient leaves too large of a deficit for bone to form on its own and requires reconstruction with metal implants or allografts. Both methods require the bone to heal, either to the implant or across the allograft junction, often in the setting of marrow-killing chemotherapy. Therefore, the issue of bone regeneration within the surgically resected margins remains an important challenge for the patient, family, and treating providers. Mesenchymal stem/stromal cells (MSCs) are potential agents for enhancing bone regeneration post tumor resection. MSCs, used with scaffolds and growth factors, show promise in fostering bone regeneration in OS cases. We spotlight two MSC types-bone marrow-derived (BM-MSCs) and adipose tissue-derived (ASCs)-highlighting their bone regrowth facilitation and immunomodulatory effects on immune cells like macrophages and T cells, enhancing therapeutic outcomes. The objective of this review is two-fold: review work demonstrating any ability of MSCs to target the deranged immune system in the OS microenvironment, and synthesize the available literature on the use of MSCs as a therapeutic option for stimulating bone regrowth in OS patients post bone resection. When it comes to repairing bone defects, both MB-MSCs and ASCs hold great potential for stimulating bone regeneration. Research has showcased their effectiveness in reconstructing bone defects while maintaining a non-tumorigenic role following wide resection of bone tumors, underscoring their capability to enhance bone healing and regeneration following tumor excisions.
Collapse
Affiliation(s)
- Max Baron
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.B.); (P.D.); (T.M.B.)
| | - Philip Drohat
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.B.); (P.D.); (T.M.B.)
| | - Brooke Crawford
- Sarcoma Biology Laboratory, Department of Orthopedics, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (B.C.); (F.J.H.)
| | - Francis J. Hornicek
- Sarcoma Biology Laboratory, Department of Orthopedics, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (B.C.); (F.J.H.)
| | - Thomas M. Best
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.B.); (P.D.); (T.M.B.)
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.B.); (P.D.); (T.M.B.)
- Diabetes Research Institute, Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
26
|
Anastasio AT, Bagheri K, Adams SB. Contemporary Review: The Use of Adipocyte-Derived Mesenchymal Stem Cells in Pathologies of the Foot and Ankle. FOOT & ANKLE ORTHOPAEDICS 2023; 8:24730114231207643. [PMID: 37929076 PMCID: PMC10623921 DOI: 10.1177/24730114231207643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023] Open
Affiliation(s)
| | - Kian Bagheri
- Department of Orthopedic Surgery, Duke University Hospital, Durham, NC, USA
- Campbell University School of Osteopathic Medicine, Lillington, NC, USA
| | - Samuel B. Adams
- Department of Orthopedic Surgery, Duke University Hospital, Durham, NC, USA
| |
Collapse
|
27
|
Quintero Sierra LA, Biswas R, Busato A, Conti A, Ossanna R, Conti G, Zingaretti N, Caputo M, Cuppari C, Parodi PC, Sbarbati A, Riccio M, De Francesco F. In Vitro Study of a Novel Vibrio alginolyticus-Based Collagenase for Future Medical Application. Cells 2023; 12:2025. [PMID: 37626834 PMCID: PMC10453626 DOI: 10.3390/cells12162025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/28/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Mesenchymal stem cells extracted from adipose tissue are particularly promising given the ease of harvest by standard liposuction and reduced donor site morbidity. This study proposes a novel enzymatic method for isolating stem cells using Vibrio alginolyticus collagenase, obtaining a high-quality product in a reduced time. Initially, the enzyme concentration and incubation time were studied by comparing cellular yield, proliferation, and clonogenic capacities. The optimized protocol was phenotypically characterized, and its ability to differentiate in the mesodermal lineages was evaluated. Subsequently, that protocol was compared with two Clostridium histolyticum-based collagenases, and other tests for cellular integrity were performed to evaluate the enzyme's effect on expanded cells. The best results showed that using a concentration of 3.6 mg/mL Vibrio alginolyticus collagenase allows extracting stem cells from adipose tissue after 20 min of enzymatic reaction like those obtained with Clostridium histolyticum-based collagenases after 45 min. Moreover, the extracted cells with Vibrio alginolyticus collagenase presented the phenotypic characteristics of stem cells that remain after culture conditions. Finally, it was seen that Vibrio alginolyticus collagenase does not reduce the vitality of expanded cells as Clostridium histolyticum-based collagenase does. These findings suggest that Vibrio alginolyticus collagenase has great potential in regenerative medicine, given its degradation selectivity by protecting vital structures for tissue restructuration.
Collapse
Affiliation(s)
- Lindsey Alejandra Quintero Sierra
- Department of Neuroscience, Biomedicine, and Movement Sciences, Human Anatomy and Histology Section, University of Verona, 37134 Verona, Italy; (L.A.Q.S.); (R.B.); (A.B.); (A.C.); (R.O.); (G.C.); (A.S.)
| | - Reetuparna Biswas
- Department of Neuroscience, Biomedicine, and Movement Sciences, Human Anatomy and Histology Section, University of Verona, 37134 Verona, Italy; (L.A.Q.S.); (R.B.); (A.B.); (A.C.); (R.O.); (G.C.); (A.S.)
| | - Alice Busato
- Department of Neuroscience, Biomedicine, and Movement Sciences, Human Anatomy and Histology Section, University of Verona, 37134 Verona, Italy; (L.A.Q.S.); (R.B.); (A.B.); (A.C.); (R.O.); (G.C.); (A.S.)
| | - Anita Conti
- Department of Neuroscience, Biomedicine, and Movement Sciences, Human Anatomy and Histology Section, University of Verona, 37134 Verona, Italy; (L.A.Q.S.); (R.B.); (A.B.); (A.C.); (R.O.); (G.C.); (A.S.)
| | - Riccardo Ossanna
- Department of Neuroscience, Biomedicine, and Movement Sciences, Human Anatomy and Histology Section, University of Verona, 37134 Verona, Italy; (L.A.Q.S.); (R.B.); (A.B.); (A.C.); (R.O.); (G.C.); (A.S.)
| | - Giamaica Conti
- Department of Neuroscience, Biomedicine, and Movement Sciences, Human Anatomy and Histology Section, University of Verona, 37134 Verona, Italy; (L.A.Q.S.); (R.B.); (A.B.); (A.C.); (R.O.); (G.C.); (A.S.)
| | - Nicola Zingaretti
- Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy; (N.Z.); (P.C.P.)
| | - Michele Caputo
- Fidia Farmaceutici S.p.A., R&D Local Unit Fidia Research Sud, Contrada Pizzuta, 96017 Noto, Italy (C.C.)
| | - Christian Cuppari
- Fidia Farmaceutici S.p.A., R&D Local Unit Fidia Research Sud, Contrada Pizzuta, 96017 Noto, Italy (C.C.)
| | - Pier Camillo Parodi
- Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, Department of Medical Area (DAME), University of Udine, 33100 Udine, Italy; (N.Z.); (P.C.P.)
- Research and Training Center in Regenerative Surgery, Accademia del Lipofilling, 61025 Montelabbate (PU), Italy
| | - Andrea Sbarbati
- Department of Neuroscience, Biomedicine, and Movement Sciences, Human Anatomy and Histology Section, University of Verona, 37134 Verona, Italy; (L.A.Q.S.); (R.B.); (A.B.); (A.C.); (R.O.); (G.C.); (A.S.)
- Research and Training Center in Regenerative Surgery, Accademia del Lipofilling, 61025 Montelabbate (PU), Italy
| | - Michele Riccio
- Research and Training Center in Regenerative Surgery, Accademia del Lipofilling, 61025 Montelabbate (PU), Italy
- Department of Reconstructive Surgery and Hand Surgery, AOU “Ospedali Riuniti”, 60126 Ancona, Italy
| | - Francesco De Francesco
- Department of Reconstructive Surgery and Hand Surgery, AOU “Ospedali Riuniti”, 60126 Ancona, Italy
| |
Collapse
|
28
|
Li Y, Shi G, Liang W, Shang H, Li H, Han Y, Zhao W, Bai L, Qin C. Allogeneic Adipose-Derived Mesenchymal Stem Cell Transplantation Alleviates Atherosclerotic Plaque by Inhibiting Ox-LDL Uptake, Inflammatory Reaction and Endothelial Damage in Rabbits. Cells 2023; 12:1936. [PMID: 37566014 PMCID: PMC10417209 DOI: 10.3390/cells12151936] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/15/2023] [Accepted: 07/19/2023] [Indexed: 08/12/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease of arteries fueled by lipids. It is a major cause of cardiovascular morbidity and mortality. Mesenchymal stem cells have been used for the treatment of atherosclerotic lesions. Adipose-derived stem cells (ADSCs) have been shown to regulate the activation state of macrophages and exhibit anti-inflammatory capabilities. However, the effect of allogeneic ADSCs in the treatment of AS have not been investigated. In this study, the early treatment effect and preliminary mechanism analysis of allogeneic rabbit ADSCs intravenous transplantation were investigated in a high-fat diet rabbit model. The polarization mechanism of rabbit ADSCs on the macrophage was further analyzed in vitro. Compared with the model group, blood lipid levels declined, the plaque area, oxidized low-density lipoprotein (ox-LDL) uptake, scavenger receptor A1 and cluster of differentiation (CD) 36 levels were all significantly reduced, and the accumulation of inflammatory M1 macrophages, apoptosis, interleukin (IL)-6 and tumor necrosis factor (TNF)-α expression were decreased. The endothelial cells (CD31), M2 macrophages, IL-10 and the transforming growth factor (TGF)-β levels increased. In vitro, ADSCs can promote the M1 macrophage phenotypic switch toward the M2 macrophage through their secreted exosomes, and the main mechanism includes increasing arginase 1 expression and IL-10 secretion, declining inducible nitric oxide synthase (iNOS) expression and TNF-α secretion, and activating the STAT6 pathway. Therefore, allogeneic rabbit ADSC transplantation can transmigrate to the aortic atherosclerotic plaques and show a good effect in lowering blood lipids and alleviating atherosclerotic plaque in the early stage of AS by inhibiting ox-LDL uptake, inflammatory response, and endothelial damage.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Chuan Qin
- NHC Key Laboratory of Human Diseases Comparative Medicine, National Human Diseases Animal Model Resource Center, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Medical Laboratory Animal Science, Chinese Academy of Medical Science (CAMS) & Comparative Medicine Centre, Peking Union Medical College (PUMC), Beijing 100021, China
| |
Collapse
|
29
|
Russo E, Corrao S, Di Gaudio F, Alberti G, Caprnda M, Kubatka P, Kruzliak P, Miceli V, Conaldi PG, Borlongan CV, La Rocca G. Facing the Challenges in the COVID-19 Pandemic Era: From Standard Treatments to the Umbilical Cord-Derived Mesenchymal Stromal Cells as a New Therapeutic Strategy. Cells 2023; 12:1664. [PMID: 37371134 PMCID: PMC10297457 DOI: 10.3390/cells12121664] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/10/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), the pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which counts more than 650 million cases and more than 6.6 million of deaths worldwide, affects the respiratory system with typical symptoms such as fever, cough, sore throat, acute respiratory distress syndrome (ARDS), and fatigue. Other nonpulmonary manifestations are related with abnormal inflammatory response, the "cytokine storm", that could lead to a multiorgan disease and to death. Evolution of effective vaccines against SARS-CoV-2 provided multiple options to prevent the infection, but the treatment of the severe forms remains difficult to manage. The cytokine storm is usually counteracted with standard medical care and anti-inflammatory drugs, but researchers moved forward their studies on new strategies based on cell therapy approaches. The perinatal tissues, such as placental membranes, amniotic fluid, and umbilical cord derivatives, are enriched in mesenchymal stromal cells (MSCs) that exert a well-known anti-inflammatory role, immune response modulation, and tissue repair. In this review, we focused on umbilical-cord-derived MSCs (UC-MSCs) used in in vitro and in vivo studies in order to evaluate the weakening of the severe symptoms, and on recent clinical trials from different databases, supporting the favorable potential of UC-MSCs as therapeutic strategy.
Collapse
Affiliation(s)
- Eleonora Russo
- Section of Histology and Embryology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| | - Simona Corrao
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per per i Trapianti e Terapie Ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | | | - Giusi Alberti
- Section of Histology and Embryology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| | - Martin Caprnda
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University, University Hospital Bratislava, 81499 Bratislava, Slovakia;
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03649 Martin, Slovakia;
| | - Peter Kruzliak
- Research and Development Services, Pradlacka 18, 61300 Brno, Czech Republic;
| | - Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per per i Trapianti e Terapie Ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per per i Trapianti e Terapie Ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | - Cesario Venturina Borlongan
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Giampiero La Rocca
- Section of Histology and Embryology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| |
Collapse
|
30
|
Melzer M, Burk J, Guest DJ, Dudhia J. Influence of Rho/ROCK inhibitor Y-27632 on proliferation of equine mesenchymal stromal cells. Front Vet Sci 2023; 10:1154987. [PMID: 37346276 PMCID: PMC10279950 DOI: 10.3389/fvets.2023.1154987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
Mesenchymal stromal cells (MSC) isolated form bone marrow and adipose tissue are the most common cells used for cell therapy of orthopedic diseases. MSC derived from different tissues show differences in terms of their proliferation, differentiation potential and viability in prolonged cell culture. This suggests that there may be subtle differences in intracellular signaling pathways that modulate these cellular characteristics. The Rho/ROCK signaling pathway is essential for many cellular functions. Targeting of this pathway by the ROCK inhibitor Y-27632 has been shown to be beneficial for cell viability and proliferation of different cell types. The aim of this study was to investigate the effects of Rho/ROCK inhibition on equine MSC proliferation using bone marrow-derived MSC (BMSC) and adipose-derived MSC (ASC). Primary ASC and BMSC were stimulated with or without 10 ng/mL TGF-β3 or 10 μM Y-27632, as well as both in combination. Etoposide at 10 μM was used as a positive control for inhibition of cell proliferation. After 48 h of stimulation, cell morphology, proliferation activity and gene expression of cell senescence markers p53 and p21 were assessed. ASC showed a trend for higher basal proliferation than BMSC, which was sustained following stimulation with TGF-β3. This included a higher proliferation with TGF-β3 stimulation compared to Y-27632 stimulation (p < 0.01), but not significantly different to the no treatment control when used in combination. Expression of p21 and p53 was not altered by stimulation with TGF-β3 and/or Y-27632 in either cell type. In summary, the Rho/ROCK inhibitor Y-27632 had no effect on proliferation activity and did not induce cell senescence in equine ASC and BMSC.
Collapse
Affiliation(s)
- Michaela Melzer
- Equine Clinic (Surgery, Orthopedics), Faculty of Veterinary Medicine, Justus Liebig University, Giessen, Germany
| | - Janina Burk
- Equine Clinic (Surgery, Orthopedics), Faculty of Veterinary Medicine, Justus Liebig University, Giessen, Germany
| | - Deborah J. Guest
- Department of Clinical Sciences and Services, Royal Veterinary College, Hertfordshire, United Kingdom
| | - Jayesh Dudhia
- Department of Clinical Sciences and Services, Royal Veterinary College, Hertfordshire, United Kingdom
| |
Collapse
|
31
|
Yue M, Liu Y, Zhang P, Li Z, Zhou Y. Integrative Analysis Reveals the Diverse Effects of 3D Stiffness upon Stem Cell Fate. Int J Mol Sci 2023; 24:9311. [PMID: 37298263 PMCID: PMC10253631 DOI: 10.3390/ijms24119311] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/09/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
The origin of life and native tissue development are dependent on the heterogeneity of pluripotent stem cells. Bone marrow mesenchymal stem cells (BMMSCs) are located in a complicated niche with variable matrix stiffnesses, resulting in divergent stem cell fates. However, how stiffness drives stem cell fate remains unknown. For this study, we performed whole-gene transcriptomics and precise untargeted metabolomics sequencing to elucidate the complex interaction network of stem cell transcriptional and metabolic signals in extracellular matrices (ECMs) with different stiffnesses, and we propose a potential mechanism involved in stem cell fate decision. In a stiff (39~45 kPa) ECM, biosynthesis of aminoacyl-tRNA was up-regulated, and increased osteogenesis was also observed. In a soft (7~10 kPa) ECM, biosynthesis of unsaturated fatty acids and deposition of glycosaminoglycans were increased, accompanied by enhanced adipogenic/chondrogenic differentiation of BMMSCs. In addition, a panel of genes responding to the stiffness of the ECM were validated in vitro, mapping out the key signaling network that regulates stem cells' fate decisions. This finding of "stiffness-dependent manipulation of stem cell fate" provides a novel molecular biological basis for development of potential therapeutic targets within tissue engineering, from both a cellular metabolic and a biomechanical perspective.
Collapse
Affiliation(s)
- Muxin Yue
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Zheng Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| |
Collapse
|
32
|
Petrova V, Vachkova E. Outlook of Adipose-Derived Stem Cells: Challenges to Their Clinical Application in Horses. Vet Sci 2023; 10:vetsci10050348. [PMID: 37235430 DOI: 10.3390/vetsci10050348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Adipose tissue is recognized as the major endocrine organ, potentially acting as a source of mesenchymal stem cells for various applications in regenerative medicine. Athletic horses are often exposed to traumatic injuries, resulting in severe financial losses. The development of adipose-derived stem cells' regenerative potential depends on many factors. The extraction of stem cells from subcutaneous adipose tissue is non-invasive, non-traumatic, cheaper, and safer than other sources. Since there is a lack of unique standards for identification, the isolated cells and applied differentiation protocols are often not species-specific; therefore, the cells cannot reveal their multipotent properties, so their stemness features remain questionable. The current review discusses some aspects of the specificity of equine adipose stem cells concerning their features, immunophenotyping, secretome profile, differentiation abilities, culturing conditions, and consequent possibilities for clinical application in concrete disorders. The presented new approaches elucidate the possibility of the transition from cell-based to cell-free therapy with regenerative purposes in horses as an alternative treatment to cellular therapy. In conclusion, their clinical benefits should not be underestimated due to the higher yield and the physiological properties of adipose-derived stem cells that facilitate the healing and tissue regeneration process and the ability to amplify the effects of traditional treatments. More profound studies are necessary to apply these innovative approaches when treating traumatic disorders in racing horses.
Collapse
Affiliation(s)
- Valeria Petrova
- Department of Pharmacology, Animal Physiology and Physiological Chemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria
| | - Ekaterina Vachkova
- Department of Pharmacology, Animal Physiology and Physiological Chemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria
| |
Collapse
|
33
|
Lischer M, di Summa PG, Petrou IG, Schaefer DJ, Guzman R, Kalbermatten DF, Madduri S. Mesenchymal Stem Cells in Nerve Tissue Engineering: Bridging Nerve Gap Injuries in Large Animals. Int J Mol Sci 2023; 24:ijms24097800. [PMID: 37175506 PMCID: PMC10177884 DOI: 10.3390/ijms24097800] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Cell-therapy-based nerve repair strategies hold great promise. In the field, there is an extensive amount of evidence for better regenerative outcomes when using tissue-engineered nerve grafts for bridging severe gap injuries. Although a massive number of studies have been performed using rodents, only a limited number involving nerve injury models of large animals were reported. Nerve injury models mirroring the human nerve size and injury complexity are crucial to direct the further clinical development of advanced therapeutic interventions. Thus, there is a great need for the advancement of research using large animals, which will closely reflect human nerve repair outcomes. Within this context, this review highlights various stem cell-based nerve repair strategies involving large animal models such as pigs, rabbits, dogs, and monkeys, with an emphasis on the limitations and strengths of therapeutic strategy and outcome measurements. Finally, future directions in the field of nerve repair are discussed. Thus, the present review provides valuable knowledge, as well as the current state of information and insights into nerve repair strategies using cell therapies in large animals.
Collapse
Affiliation(s)
- Mirko Lischer
- Center for Bioengineering and Regenerative Medicine, Department of Biomedical Engineering, University of Basel, 4123 Allschwil, Switzerland
| | - Pietro G di Summa
- Department of Plastic, Reconstructive and Hand Surgery, University Hospital of Lausanne and University of Lausanne, 1015 Lausanne, Switzerland
| | - Ilias G Petrou
- Plastic, Reconstructive and Aesthetic Surgery, Department of Surgery, University Hospitals and University of Geneva, 1205 Geneva, Switzerland
| | - Dirk J Schaefer
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Raphael Guzman
- Department of Neurosurgery, University Hospital Basel, 4031 Basel, Switzerland
| | - Daniel F Kalbermatten
- Plastic, Reconstructive and Aesthetic Surgery, Department of Surgery, University Hospitals and University of Geneva, 1205 Geneva, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Srinivas Madduri
- Center for Bioengineering and Regenerative Medicine, Department of Biomedical Engineering, University of Basel, 4123 Allschwil, Switzerland
- Plastic, Reconstructive and Aesthetic Surgery, Department of Surgery, University Hospitals and University of Geneva, 1205 Geneva, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
- Bioengineering and Neuroregeneration, Department of Surgery, Geneva University Hospitals and University of Geneva, 1205 Geneva, Switzerland
| |
Collapse
|
34
|
Lee LM, Klarmann GJ, Haithcock DW, Wang Y, Bhatt KH, Prabhakarpandian B, Pant K, Alvarez LM, Lai E. Label-free enrichment of human adipose-derived stem cells using a continuous microfluidic sorting cascade. LAB ON A CHIP 2023; 23:2131-2140. [PMID: 36974599 DOI: 10.1039/d2lc01138g] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Human adipose tissue is a rich source of mesenchymal stem cells (MSCs). Human adipose-derived stem cells (ADSCs) are first prepared by tissue digestion of lipoaspirate. The remaining constituent contains a mixture of ADSCs, other cell types and lysed fragments. We have developed a scalable microfluidic sorter cascade which enabled high-throughput and label-free enrichment of ADSCs prepared from tissue-digested human adipose samples to improve the quality of purified stem cell product. The continuous microfluidic sorter cascade was composed of spiral-shaped inertial and deterministic lateral displacement (DLD) sorters which separated cells based on size difference. The cell count characterization results showed >90% separation efficiency. We also demonstrated that the enriched ADSC sub-population by the microfluidic sorter cascade yielded 6× enhancement of expansion capacity in tissue culture. The incorporation of this microfluidic sorter cascade into ADSC preparation workflow facilitates the generation of transplantation-scale stem cell product. We anticipate our stem cell microfluidic sorter cascade will find a variety of research and clinical applications in tissue engineering and regeneration medicine.
Collapse
Affiliation(s)
- Lap Man Lee
- CFD Research Corporation, 6820 Moquin Dr NW, Huntsville, AL, 35806, U.S.A.
| | - George J Klarmann
- The Geneva Foundation, 9410 Key West Ave, Rockville, MD 20850, U.S.A
| | - Dustin W Haithcock
- CFD Research Corporation, 6820 Moquin Dr NW, Huntsville, AL, 35806, U.S.A.
| | - Yi Wang
- Mechanical Engineering, College of Engineering and Computing, University of South Carolina, 300 Main Street, Columbia, SC 29208, USA
| | - Ketan H Bhatt
- CFD Research Corporation, 6820 Moquin Dr NW, Huntsville, AL, 35806, U.S.A.
| | | | - Kapil Pant
- CFD Research Corporation, 6820 Moquin Dr NW, Huntsville, AL, 35806, U.S.A.
| | - Luis M Alvarez
- Walter Reed National Military Medical Center, 4494 Palmer Rd N, Bethesda, MD 20814, U.S.A
| | - Eva Lai
- Mechanical Engineering & Materials Science, Swanson School of Engineering, University of Pittsburgh, 636 Benedum Hall, 3700 O'Hara Street, Pittsburgh, PA 15261, U.S.A
| |
Collapse
|
35
|
Ye P, Feng L, Zhang D, Li R, Wen Y, Tong X, Shi S, Dong C. Metformin Ameliorates D-Galactose-Induced Senescent Human Bone Marrow-Derived Mesenchymal Stem Cells by Enhancing Autophagy. Stem Cells Int 2023; 2023:1429642. [PMID: 37035446 PMCID: PMC10079386 DOI: 10.1155/2023/1429642] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/16/2023] [Accepted: 03/11/2023] [Indexed: 04/04/2023] Open
Abstract
Human bone marrow-derived mesenchymal stem cells (hBMSCs) are promising candidates for stem cell therapy in clinical trials. Applications of hBMSCs in clinical therapy are limited by cellular senescence due to long-term ex vivo expansion. Metformin, an oral hypoglycemic drug for type 2 diabetes, has been shown to have antiaging effects. However, the mechanisms of metformin in antiaging treatment remain controversial. Here, we used D-galactose (D-gal) to establish an appropriate model of senescent hBMSCs to explore the antiaging effects of metformin. Following metformin treatment with a low concentration range, senescence phenotypes induced by D-gal significantly changed, including generation of reactive oxygen species (ROS), loss of mitochondrial membrane potential (MMP), and cell cycle arrest. In contrast, no apparent change was found in unsenescent hBMSCs. Furthermore, the results show that activation of 5'AMP-activated protein kinase (AMPK) by metformin enhances cell autophagy in senescent hBMSCs. These findings suggest that metformin exerts antiaging function within the low concentration range by enhancing autophagy and exhibits potential benefits for clinical stem cell therapy by ameliorating the ex vivo replicative senescence of hBMSCs.
Collapse
Affiliation(s)
- Pingting Ye
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| | - Lei Feng
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| | - Dan Zhang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| | - Ruihao Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| | - Yixuan Wen
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| | - Xiaohan Tong
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| | - Shuo Shi
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| | - Chunyan Dong
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| |
Collapse
|
36
|
Giorgino R, Albano D, Fusco S, Peretti GM, Mangiavini L, Messina C. Knee Osteoarthritis: Epidemiology, Pathogenesis, and Mesenchymal Stem Cells: What Else Is New? An Update. Int J Mol Sci 2023; 24:ijms24076405. [PMID: 37047377 PMCID: PMC10094836 DOI: 10.3390/ijms24076405] [Citation(s) in RCA: 143] [Impact Index Per Article: 71.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
Osteoarthritis (OA) is a chronic disease and the most common orthopedic disorder. A vast majority of the social OA burden is related to hips and knees. The prevalence of knee OA varied across studies and such differences are reflected by the heterogeneity of data reported by studies conducted worldwide. A complete understanding of the pathogenetic mechanisms underlying this pathology is essential. The OA inflammatory process starts in the synovial membrane with the activation of the immune system, involving both humoral and cellular mediators. A crucial role in this process is played by the so-called “damage-associated molecular patterns” (DAMPs). Mesenchymal stem cells (MSCs) may be a promising option among all possible therapeutic options. However, many issues are still debated, such as the best cell source, their nature, and the right amount. Further studies are needed to clarify the remaining doubts. This review provides an overview of the most recent and relevant data on the molecular mechanism of cartilage damage in knee OA, including current therapeutic approaches in regenerative medicine.
Collapse
|
37
|
Fujii Y, Hatori A, Chikazu D, Ogasawara T. Application of Dental Pulp Stem Cells for Bone and Neural Tissue Regeneration in Oral and Maxillofacial Region. Stem Cells Int 2023; 2023:2026572. [PMID: 37035445 PMCID: PMC10076122 DOI: 10.1155/2023/2026572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 10/21/2022] [Accepted: 03/18/2023] [Indexed: 03/31/2023] Open
Abstract
In the oral and maxillofacial region, the treatment of severe bone defects, caused by fractures, cancers, congenital abnormalities, etc., remains a great challenge. In addition, neurological disorders are frequently accompanied by these bone defects or the treatments for them. Therefore, novel bone regenerative techniques and methods to repair nerve injury are eagerly sought. Among them, strategies using dental pulp stem cells (DPSCs) are promising options. Human DPSCs can be collected easily from extracted teeth and are now considered a type of mesenchymal stem cell with higher clonogenic and proliferative potential. DPSCs have been getting attention as a cell source for bone and nerve regeneration. In this article, we reviewed the latest studies on osteogenic or neural differentiation of DPSCs as well as bone or neural regeneration methods using DPSCs and discussed the potential of DPSCs for bone and nerve tissue regeneration.
Collapse
|
38
|
Ozden Akkaya O, Dikmen T, Nawaz S, Kibria AG, Altunbaş K, Yağci A, Erdoğan M, Yaprakci MV. Comparison of proliferation and osteogenic differentiation potential of bovine adipose tissue and bone marrow derived stem cells. Biotech Histochem 2023; 98:267-279. [PMID: 36815431 DOI: 10.1080/10520295.2023.2177347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Abstract
Bone marrow derived stem cells (BMSC) are the most utilized cell type in the field of bone regeneration. Although BMSC are both safe and efficacious, the search for alternative sources for stem cells continues. We investigated bovine BMSC and adipose tissue derived mesenchymal stem cells (ATSC) using immunofluorescence and PCR. We further compared the osteogenic differentiation potentials of both sources of stem cells. We assessed alkaline phosphatase (ALP) enzyme levels and calcium deposition in differentiating cells at days 7, 14 and 21 to compare the osteogenic differentiation capability of both cell types. We found that ATSC expressed significantly higher ALP levels compared to BMSC throughout osteogenic differentiation. Calcium deposition was greater in ATSC than BMSC at days 7 and 14. By the end of day 21, BMSC produced greater calcium deposition. We found that ATSC undergo osteogenic differentiation more rapidly than BMSC, but BMSC provide greater mineralization over longer periods.
Collapse
Affiliation(s)
- Ozlem Ozden Akkaya
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye
| | - Tayfun Dikmen
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye
| | - Shah Nawaz
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye.,Department of Anatomy, Faculty of Veterinary Science, University of Agriculture, Faisalabad, Pakistan
| | - Asm Golam Kibria
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye.,Department of Anatomy and Histology, Chattogram University of Veterinary and Animal Sciences, Chattogram, Bangladesh
| | - Korhan Altunbaş
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye
| | - Artay Yağci
- Department of Histology and Embryology, Milas Veterinary Faculty, Mugla Sıtkı Kocman University, Mugla, Türkiye
| | - Metin Erdoğan
- Department of Veterinary Biology and Genetics, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye
| | - Mustafa Volkan Yaprakci
- Department of Surgery, Faculty of Veterinary Medicine, Afyon Kocatepe University, Afyonkarahisar, Türkiye
| |
Collapse
|
39
|
Sueters J, Groenman FA, Bouman MB, Roovers JPW, de Vries R, Smit TH, Huirne JAF. Tissue Engineering Neovagina for Vaginoplasty in Mayer-Rokitansky-Küster-Hauser Syndrome and Gender Dysphoria Patients: A Systematic Review. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:28-46. [PMID: 35819292 DOI: 10.1089/ten.teb.2022.0067] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Background: Vaginoplasty is a surgical solution to multiple disorders, including Mayer-Rokitansky-Küster-Hauser syndrome and male-to-female gender dysphoria. Using nonvaginal tissues for these reconstructions is associated with many complications, and autologous vaginal tissue may not be sufficient. The potential of tissue engineering for vaginoplasty was studied through a systematic bibliography search. Cell types, biomaterials, and signaling factors were analyzed by investigating advantages, disadvantages, complications, and research quantity. Search Methods: A systematic search was performed in Medline, EMBASE, Web of Science, and Scopus until March 8, 2022. Term combinations for tissue engineering, guided tissue regeneration, regenerative medicine, and tissue scaffold were applied, together with vaginoplasty and neovagina. The snowball method was performed on references and a Google Scholar search on the first 200 hits. Original research articles on human and/or animal subjects that met the inclusion (reconstruction of vaginal tissue and tissue engineering method) and no exclusion criteria (not available as full text; written in foreign language; nonoriginal study article; genital surgery other than neovaginal reconstruction; and vaginal reconstruction with autologous or allogenic tissue without tissue engineering or scaffold) were assessed. The Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) checklist, the Newcastle-Ottawa Scale, and the Gold Standard Publication Checklist were used to evaluate article quality and bias. Outcomes: A total of 31 out of 1569 articles were included. Data extraction was based on cell origin and type, biomaterial nature and composition, host species, number of hosts and controls, neovaginal size, replacement fraction, and signaling factors. An overview of used tissue engineering methods for neovaginal formation was created, showing high variance of cell types, biomaterials, and signaling factors and the same topics were rarely covered multiple times. Autologous vaginal cells and extracellular matrix-based biomaterials showed preferential properties, and stem cells carry potential. However, quality confirmation of orthotopic cell-seeded acellular vaginal matrix by clinical trials is needed as well as exploration of signaling factors for vaginoplasty. Impact statement General article quality was weak to sufficient due to unreported cofounders and incomplete animal study descriptions. Article quality and heterogenicity made identification of optimal cell types, biomaterials, or signaling factors unreliable. However, trends showed that autologous cells prevent complications and compatibility issues such as healthy cell destruction, whereas stem cells prevent cross talk (interference of signaling pathways by signals from other cell types) and rejection (but need confirmation testing beyond animal trials). Natural (orthotopic) extracellular matrix biomaterials have great preferential properties that encourage future research, and signaling factors for vascularization are important for tissue engineering of full-sized neovagina.
Collapse
Affiliation(s)
- Jayson Sueters
- Department of Gynaecology and Amsterdam Reproduction and Development, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Freek A Groenman
- Department of Obstetrics and Gynecology, Amsterdam Reproduction and Development, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.,Centre of Expertise on Gender Dysphoria, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Mark-Bram Bouman
- Centre of Expertise on Gender Dysphoria, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Jan Paul W Roovers
- Department of Obstetrics and Gynecology, Amsterdam Reproduction and Development, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Ralph de Vries
- Medical Library, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Theo H Smit
- Department of Gynaecology and Amsterdam Reproduction and Development, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.,Department of Medical Biology, Amsterdam UMC location AMC, Amsterdam, The Netherlands
| | - Judith A F Huirne
- Department of Gynaecology and Amsterdam Reproduction and Development, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.,Research Institute Reproduction and Development, Amsterdam UMC location AMC, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Everts PA, Panero AJ. Basic Science of Autologous Orthobiologics. Phys Med Rehabil Clin N Am 2023; 34:25-47. [DOI: 10.1016/j.pmr.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
41
|
Muthu S, Patil SC, Jeyaraman N, Jeyaraman M, Gangadaran P, Rajendran RL, Oh EJ, Khanna M, Chung HY, Ahn BC. Comparative effectiveness of adipose-derived mesenchymal stromal cells in the management of knee osteoarthritis: A meta-analysis. World J Orthop 2023; 14:23-41. [PMID: 36686284 PMCID: PMC9850793 DOI: 10.5312/wjo.v14.i1.23] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/20/2022] [Accepted: 12/13/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most common joint disorder, is associated with an increasing socioeconomic impact owing to the ageing population. AIM To analyze and compare the efficacy and safety of bone-marrow-derived mesenchymal stromal cells (BM-MSCs) and adipose tissue-derived MSCs (AD-MSCs) in knee OA management from published randomized controlled trials (RCTs). METHODS Independent and duplicate electronic database searches were performed, including PubMed, EMBASE, Web of Science, and Cochrane Library, until August 2021 for RCTs that analyzed the efficacy and safety of AD-MSCs and BM-MSCs in the management of knee OA. The visual analog scale (VAS) score for pain, Western Ontario McMaster Universities Osteoarthritis Index (WOMAC), Lysholm score, Tegner score, magnetic resonance observation of cartilage repair tissue score, knee osteoarthritis outcome score (KOOS), and adverse events were analyzed. Analysis was performed on the R-platform using OpenMeta (Analyst) software. Twenty-one studies, involving 936 patients, were included. Only one study compared the two MSC sources without patient randomization; hence, the results of all included studies from both sources were pooled, and a comparative critical analysis was performed. RESULTS At six months, both AD-MSCs and BM-MSCs showed significant VAS improvement (P = 0.015, P = 0.012); this was inconsistent at 1 year for BM-MSCs (P < 0.001, P = 0.539), and AD-MSCs outperformed BM-MSCs compared to controls in measures such as WOMAC (P < 0.001, P = 0.541), Lysholm scores (P = 0.006; P = 0.933), and KOOS (P = 0.002; P = 0.012). BM-MSC-related procedures caused significant adverse events (P = 0.003) compared to AD-MSCs (P = 0.673). CONCLUSION Adipose tissue is superior to bone marrow because of its safety and consistent efficacy in improving pain and functional outcomes. Future trials are urgently warranted to validate our findings and reach a consensus on the ideal source of MSCs for managing knee OA.
Collapse
Affiliation(s)
- Sathish Muthu
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul 624001, Tamil Nadu, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India
- Research Associate, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Indian Stem Cell Study Group Association, Lucknow 226001, Uttar Pradesh, India
| | - Sandesh C Patil
- Department of Orthopaedic Rheumatology, Dr. RML National Law University, Lucknow 226012, Uttar Pradesh, India
| | - Naveen Jeyaraman
- Indian Stem Cell Study Group Association, Lucknow 226001, Uttar Pradesh, India
- Department of Orthopaedic Rheumatology, Dr. RML National Law University, Lucknow 226012, Uttar Pradesh, India
| | - Madhan Jeyaraman
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India
- Research Associate, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Indian Stem Cell Study Group Association, Lucknow 226001, Uttar Pradesh, India
- Department of Orthopaedics, ACS Medical College & Hospital, Dr MGR Educational and Research Institute, Chennai 600056, Tamil Nadu, India
| | - Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
| | - Eun Jung Oh
- Department of Plastic and Reconstructive Surgery, CMRI, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea
| | - Manish Khanna
- Indian Stem Cell Study Group Association, Lucknow 226001, Uttar Pradesh, India
| | - Ho Yun Chung
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
- Department of Plastic and Reconstructive Surgery, CMRI, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, South Korea
| |
Collapse
|
42
|
Andrietti ALP, Durgam SS, Naumann B, Stewart M. Basal and inducible Osterix expression reflect equine mesenchymal progenitor cell osteogenic capacity. Front Vet Sci 2023; 10:1125893. [PMID: 37035801 PMCID: PMC10076790 DOI: 10.3389/fvets.2023.1125893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/28/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Mesenchymal stem cells are characterized by their capacities for extensive proliferation through multiple passages and, classically, tri-lineage differentiation along osteogenic, chondrogenic and adipogenic lineages. This study was carried out to compare osteogenesis in equine bone marrow-, synovium- and adipose-derived cells, and to determine whether osteogenic capacity is reflected in the basal expression of the critical osteogenic transcription factors Runx2 and Osterix. Methods Bone marrow, synovium and adipose tissue was collected from six healthy 2-year-old horses. Cells were isolated from these sources and expanded through two passages. Basal expression of Runx2 and Osterix was assessed in undifferentiated third passage cells, along with their response to osteogenic culture conditions. Results Bone marrow-derived cells had significantly higher basal expression of Osterix, but not Runx2. In osteogenic medium, bone-marrow cells rapidly developed dense, multicellular aggregates that stained strongly for mineral and alkaline phosphatase activity. Synovial and adipose cell cultures showed far less matrix mineralization. Bone marrow cells significantly up-regulated alkaline phosphatase mRNA expression and enzymatic activity at 7 and 14 days. Alkaline phosphatase expression and activity were increased in adipose cultures after 14 days, although these values were less than in bone marrow cultures. There was no change in alkaline phosphatase in synovial cultures. In osteogenic medium, bone marrow cultures increased both Runx2 and Osterix mRNA expression significantly at 7 and 14 days. Expression of both transcription factors did not change in synovial or adipose cultures. Discussion These results demonstrate that basal Osterix expression differs significantly in progenitor cells derived from different tissue sources and reflects the osteogenic potential of the cell populations.
Collapse
|
43
|
TEKİN Ö, UYANIKGİL Y, TAŞKIRAN D. Glukagon benzeri peptit-1'in yağ doku kaynaklı mezenkimal kök hücrelerinin kardiyomiyositlere dönüşmesi üzerindeki etkisi. EGE TIP DERGISI 2022. [DOI: 10.19161/etd.1180666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Amaç: Mezenkimal kök hücreler, çeşitli protokoller kullanılarak in vitro koşullarda kolaylıkla
kardiyomiyositlere farklılaşabilir. Ancak bu protokollerde kullanılan ajanların hücre canlılığı üzerinde bazı
olumsuz etkileri olduğu bildirilmiştir. Azasitidin mezenkimal kök hücreleri kalp kası hücrelerine
farklandırmak için kullanılmaktadır. Bu çalışmanın amacı, bir GLP-1 reseptör agonisti olan Eksenatid'in
insan yağ dokusu kaynaklı kök hücrelerinin kardiyomiyositlere farklılaşması ve canlılığı üzerindeki
etkilerini araştırmaktır.
Gereç ve Yöntem: Azasitidin ve Eksenatid'in insan yağ doku kaynaklı mezenkimal kök hücreler
üzerinde hücre canlılığı ve proliferasyonu üzerindeki etkileri ile sitotoksisite testleri yapıldı. Farklılanma
protokolü için, hücreler dört hafta boyunca Azasitidin ve Eksenatid ile inkübe edildi. Hücrelerin morfolojik
değişiklikleri izlendi ve kardiyomiyojenik farklılaşma belirteçlerinin (cTnI, GATA4 ve MYH7)
ekspresyonları immünohistokimyasal olarak değerlendirildi. Ayrıca kültürlerdeki kardiyak troponin I
(cTnI) seviyeleri enzime bağlı immünosorbent testi kullanılarak ölçüldü. Veriler, tek yönlü varyans analizi
(ANOVA) ve post-hoc testi ile değerlendirildi.
Bulgular: İnsan yağ doku kaynaklı mezenkimal kök hücreler üzerine Azasitidin uygulaması, kontrole
grubuna kıyasla hücre canlılığını önemli ölçüde azaltırken (%54.4) hücrelerin Azasitidin+Eksenatid ile
uygulaması doza bağlı bir şekilde hücre ölümünü önledi. Azasitidin ve Eksenatid uygulanan hücreler,
kardiyomiyojenik farklılaşma ile uyumlu önemli morfolojik değişiklikler ve kardiyomiyojenik belirteçlerde
artış gösterdi. Ayrı ayrı ve birlikte uygulama yapılan gruplarda cTnI seviyeleri kontrole göre anlamlı
derecede yüksek bulundu.
Sonuç: Bu bulgular GLP-1 reseptör agonisti Eksenatid'in, Azasitidin uygulamasının neden olduğu hücre
hasarını azaltarak İnsan yağ doku kaynaklı mezenkimal kök hücrelerin kardiyomiyojenik farklılaşması
üzerinde faydalı etkileri olabileceğini düşündürmektedir.
Collapse
|
44
|
Fujii S, Endo K, Matsuta S, Komori K, Sekiya I. Comparison of the yields and properties of dedifferentiated fat cells and mesenchymal stem cells derived from infrapatellar fat pads. Regen Ther 2022; 21:611-619. [DOI: 10.1016/j.reth.2022.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/07/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
|
45
|
Lenna S, Brozovich A, Hirase T, Paradiso F, Weiner BK, Taraballi F. Comparison between Cancellous Trabecular and Cortical Specimens from Human Lumbar Spine Samples as an Alternative Source of Mesenchymal Stromal Cells. Stem Cells Dev 2022; 31:672-683. [PMID: 36039931 DOI: 10.1089/scd.2022.0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Due to their immunosuppressive potential and ability to differentiate into multiple musculoskeletal cell lineages, mesenchymal stromal cells (MSCs) became popular in clinical trials for the treatment of musculoskeletal disorders. The aim of this study was to isolate and characterize native populations of MSCs from human cortical and cancellous bone from the posterior elements of the lumbar spine and determine what source of MSCs yield better quality and quantity of cells to be potentially use for spinal fusion repair. We were able to show that MSCs from trabecular and cortical spine had the typical MSC morphology and expression markers; the ability to differentiate in adipocyte, chondrocyte, or osteoblast but they did not have a consistent pattern in the expression of the specific differentiation lineage genes. Moreover, MSCs from both sites demonstrated an immune suppression profile suggesting that these cells may have a more promising success in applications related to immunomodulation more than exploring their ability to drive osteogenesis to prevent nonunion in spine fusion procedures.
Collapse
Affiliation(s)
- Stefania Lenna
- Houston Methodist Research Institute, Houston, Texas, United States;
| | - Ava Brozovich
- Houston Methodist Academic Institute, Houston, Texas, United States;
| | - Takashi Hirase
- Houston Methodist Orthopedics & Sports Medicine Texas Medical Center, Houston, Texas, United States;
| | | | - Bradley K Weiner
- The Houston Methodist Research Institute, Department of Nanomedicine, Houston, Texas, United States.,Houston Methodist Hospital, Department of Orthopedic Surgery, Houston, Texas, United States;
| | | |
Collapse
|
46
|
Safoine M, Côté A, Leloup R, Hayward CJ, Plourde Campagna MA, Ruel J, Fradette J. Engineering naturally-derived human connective tissues for clinical applications using a serum-free production system. Biomed Mater 2022; 17. [PMID: 35950736 DOI: 10.1088/1748-605x/ac84b9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 07/27/2022] [Indexed: 11/12/2022]
Abstract
The increasing need for tissue substitutes in reconstructive surgery spurs the development of engineering methods suited for clinical applications. Cell culture and tissue production traditionally require the use of fetal bovine serum (FBS) which is associated with various complications especially from a translational perspective. Using the self-assembly approach of tissue engineering, we hypothesized that all important parameters of tissue reconstruction can be maintained in a production system devoid of FBS from cell extraction to tissue reconstruction. We studied two commercially available serum-free medium (SFM) and xenogen-free serum-free medium (XSFM) for their impact on tissue reconstruction using human adipose-derived stem/stromal cells (ASCs) in comparison to serum-containing medium. Both media allowed higher ASC proliferation rates in primary cultures over five passages compared with 10% FBS supplemented medium while maintaining high expression of mesenchymal cell markers. For both media, we evaluated extracellular matrix production and deposition necessary to engineer manipulatable tissues using the self-assembly approach. Tissues produced in SFM exhibited a significantly increased thickness (up to 6.8-fold) compared with XSFM and FBS-containing medium. A detailed characterization of tissues produced under SFM conditions showed a substantial 50% reduction of production time without compromising key tissue features such as thickness, mechanical resistance and pro-angiogenic secretory capacities (plasminogen activator inhibitor 1, hepatocyte growth factor, vascular endothelial growth factor, angiopoietin-1) when compared to tissues produced in the control FBS-containing medium. Furthermore, we compared ASCs to the frequently used human dermal fibroblasts (DFs) in the SFM culture system. ASC-derived tissues displayed a 2.4-fold increased thickness compared to their DFs counterparts. In summary, we developed all-natural human substitutes using a production system compatible with clinical requirements. Under culture conditions devoid of bovine serum, the resulting engineered tissues displayed similar and even superior structural and functional properties over the classic FBS-containing culture conditions with a considerable 50% shortening of production time.
Collapse
Affiliation(s)
- Meryem Safoine
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada.,Division of Regenerative Medicine, CHU de Québec-Université Laval Research Centre, Québec, QC, Canada
| | - Alexandra Côté
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada.,Division of Regenerative Medicine, CHU de Québec-Université Laval Research Centre, Québec, QC, Canada
| | - Romane Leloup
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada.,Division of Regenerative Medicine, CHU de Québec-Université Laval Research Centre, Québec, QC, Canada
| | - Cindy Jean Hayward
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada.,Division of Regenerative Medicine, CHU de Québec-Université Laval Research Centre, Québec, QC, Canada
| | - Marc-André Plourde Campagna
- Bureau de design, Department of Mechanical Engineering, Faculty of Science and Engineering, Université Laval, Québec, QC, Canada
| | - Jean Ruel
- Division of Regenerative Medicine, CHU de Québec-Université Laval Research Centre, Québec, QC, Canada.,Bureau de design, Department of Mechanical Engineering, Faculty of Science and Engineering, Université Laval, Québec, QC, Canada
| | - Julie Fradette
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada.,Division of Regenerative Medicine, CHU de Québec-Université Laval Research Centre, Québec, QC, Canada.,Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
47
|
Camacho-Alonso F, Tudela-Mulero MR, Navarro JA, Buendía AJ, Mercado-Díaz AM. Use of buccal fat pad-derived stem cells cultured on bioceramics for repair of critical-sized mandibular defects in healthy and osteoporotic rats. Clin Oral Investig 2022; 26:5389-5408. [PMID: 35524820 PMCID: PMC9381637 DOI: 10.1007/s00784-022-04506-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/14/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To compare new bone formation in mandibular symphysis critical-sized bone defects (CSBDs) in healthy and osteoporotic rats filled with bioceramics (BCs) with or without buccal fat pad mesenchymal stem cells (BFPSCs). MATERIALS AND METHODS Thirty-two adult female Sprague-Dawley rats were randomized to two groups (n = 16 per group): group 1 healthy and group 2 osteoporotic (with bilateral ovariectomy). The central portion of the rat mandibular symphysis was used as a physiological CSBD. In each group, eight defects were filled with BC (hydroxyapatite 60% and β-tricalcium phosphate 40%) alone and eight with BFPSCs cultured on BC. The animals were sacrificed at 4 and 8 weeks, and the mandibles were processed for micro-computed tomography to analyze radiological union and bone mineral density (BMD); histological analysis of the bone union; and immunohistochemical analysis, which included immunoreactivity of vascular endothelial growth factor (VEGF) and bone morphogenetic protein 2 (BMP-2). RESULTS In both groups, CSBDs filled with BC + BFPSCs showed greater radiological bone union, BMD and histological bone union, and more VEGF and BMP-2 positivity, compared with CSBDs treated with BC alone at 4 and 8 weeks. CONCLUSIONS The application of BFPSCs cultured on BCs improves bone regeneration in CSBDs compared with BCs alone in healthy and osteoporotic rats. CLINICAL RELEVANCE Our results may aid bone regeneration of maxillofacial CSBDs of both healthy and osteoporotic patients, but further studies are necessary.
Collapse
Affiliation(s)
- Fabio Camacho-Alonso
- Department of Oral Surgery, University of Murcia, Murcia, Spain.
- Oral Surgery Teaching Unit, University Dental Clinic, Morales Meseguer Hospital (2Nd Floor), Marqués de los Vélez s/n, 30008, Murcia, Spain.
| | | | - J A Navarro
- Department of Histology and Pathological Anatomy, University of Murcia, Murcia, Spain
| | - A J Buendía
- Department of Histology and Pathological Anatomy, University of Murcia, Murcia, Spain
| | | |
Collapse
|
48
|
Karadag Sari EC, Ovali E. Factors affecting cell viability and the yield of adipose-derived stromal vascular fraction. J Plast Surg Hand Surg 2022; 56:249-254. [PMID: 35819816 DOI: 10.1080/2000656x.2022.2097250] [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] [Indexed: 10/17/2022]
Abstract
The stromal vascular fraction (SVF) is isolated from adipose tissue and has tremendous regenerative potential for proliferation and differentiation. This study aimed to investigate the factors affecting the cell yield and viability of the SVF to improve the outcomes of its clinical applications and enhance its clinical usage. We performed a retrospective analysis with 121 patients who underwent liposuction to harvest adipose-derived SVF. We recorded patient demographic and clinical characteristics, including age, sex, body mass index (BMI), blood type, medical comorbidities, and smoking and alcohol consumption. As for operative variables, we noted the amount of lipoaspirate and the donor areas, including the lower and entire abdomen. The viability and the cell count of SVF were documented. Sex was a statistically significant factor for viability rate (p < 0.015) and cell count (p < 0.009). Men had higher viability, while women had higher cell counts. We found a statistically significant difference in the presence of hypertension (p = 0.024) and alcohol consumption (p = 0.024). There was a statistically significant relationship between cell count and age (p < 0.001), BMI (p = 0.006), and amount of lipoaspirate (p < 0.001). Sex had significant associations with cell count and viability, while age, BMI, and lipoaspirate amount were significantly associated with cell count. Hypertension and alcohol consumption significantly affected cell count, which is the first such report of this association. Surgeons could apply this knowledge to patient selection for optimal treatment outcomes. Additionally, understanding these factors can help manage patient expectations.
Collapse
Affiliation(s)
- E Cigdem Karadag Sari
- Plastic, Reconstructive and Aesthetic Surgery, Acibadem University, School of Medicine, Acibadem Altunizade Hospital, Istanbul, Turkey
| | - Ercument Ovali
- Hematology, Acibadem University, School of Medicine, Acibadem Labcell Cellular Therapy Facility, Istanbul, Turkey
| |
Collapse
|
49
|
Yu P, Guo J, Li J, Shi X, Xu N, Jiang Y, Chen W, Hu Q. lncRNA-H19 in Fibroblasts Promotes Wound Healing in Diabetes. Diabetes 2022; 71:1562-1578. [PMID: 35472819 DOI: 10.2337/db21-0724] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022]
Abstract
Cutaneous wound healing in diabetes is impaired and would develop into nonhealing ulcerations. However, the molecular mechanism underlying the wound-healing process remains largely obscure. Here, we found that cutaneous PDGFRα+ fibroblast-expressing lncRNA-H19 (lncH19) accelerates the wound-healing process via promoting dermal fibroblast proliferation and macrophage infiltration in injured skin. PDGFRα+ cell-derived lncH19, which is lower in contents in the wound-healing cutaneous tissue of patients and mice with type 2 diabetes, is required for wound healing through promoting proliferative capacity of dermis fibroblasts as well as macrophage recruitments. Mechanistically, lncH19 relieves the cell cycle arrest of fibroblasts and increases macrophage infiltration in injured tissues via inhibiting p53 activity and GDF15 releasement. Furthermore, exosomes derived from adipocyte progenitor cells efficiently restore the impaired diabetic wound healing via delivering lncH19 to injured tissue. Therefore, our study reveals a new role for lncRNA in regulating cutaneous tissue repair and provides a novel promising insight for developing clinical treatment of diabetes.
Collapse
Affiliation(s)
- Pijun Yu
- Department of Plastic and Cosmetic Surgery, Shanghai Eighth People's Hospital, Shanghai, China
| | - Jian Guo
- Department of Plastic and Cosmetic Surgery, Shanghai Eighth People's Hospital, Shanghai, China
| | - Junjie Li
- Department of Plastic and Cosmetic Surgery, Shanghai Eighth People's Hospital, Shanghai, China
| | - Xiao Shi
- Department of Plastic and Cosmetic Surgery, Shanghai Eighth People's Hospital, Shanghai, China
| | - Ning Xu
- Department of Plastic and Cosmetic Surgery, Shanghai Eighth People's Hospital, Shanghai, China
| | - Yongkang Jiang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Chen
- Department of Plastic and Cosmetic Surgery, Shanghai Eighth People's Hospital, Shanghai, China
| | - Qin Hu
- Department of Gynecology, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
50
|
Fujita M, Matsumoto T, Hayashi S, Hashimoto S, Nakano N, Maeda T, Kuroda Y, Takashima Y, Kikuchi K, Anjiki K, Ikuta K, Onoi Y, Tachibana S, Matsushita T, Iwaguro H, Sobajima S, Hiranaka T, Kuroda R. Paracrine effect of the stromal vascular fraction containing M2 macrophages on human chondrocytes through the Smad2/3 signaling pathway. J Cell Physiol 2022; 237:3627-3639. [PMID: 35766589 DOI: 10.1002/jcp.30823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 05/27/2022] [Accepted: 06/17/2022] [Indexed: 11/08/2022]
Abstract
The adipose-derived stromal vascular fraction (SVF) is composed of a heterogeneous mix of adipose-derived stem cells (ADSCs), macrophages, pericytes, fibroblasts, blood, and other cells. Previous studies have found that the paracrine effects of SVF cells may be therapeutic, but their role in osteoarthritis treatment remains unclear. This study aimed to investigate the therapeutic effect of SVF cells on chondrocytes. Chondrocytes were seeded on culture plates alone (control) or cocultured with SVF or ADSCs on cell culture inserts. After 48 h of coculture, chondrocyte collagen II, tissue inhibitors of metalloproteinases-3 (TIMP-3), and matrix metalloproteinases-13 (MMP-13) messenger RNA (mRNA) expression levels were evaluated using reverse-transcription polymerase chain reaction, and the transforming growth factor-β (TGF-β) levels in the supernatant were measured using ELISA. Immunohistochemical staining and flow cytometry were used to evaluate the macrophages in the SVF. These macrophages were characterized according to phenotype using the F4/80, CD86, and CD163 markers. To determine whether the Smad2/3 signaling pathways were involved, the chondrocytes were pre-treated with a Smad2/3 phosphorylation inhibitor and stimulated with the SVF, and then Smad2/3 phosphorylation levels were analyzed using western blot. The mRNA expression levels of various paracrine factors and chondrocyte pellet size were also assessed. Collagen II and TIMP-3 expression were higher in the SVF group than in the ADSC group and controls, while MMP-13 expression was the highest in the ADSC group and the lowest in the controls. TGF-β levels in the SVF group were also elevated. Immunohistochemical staining and flow cytometry revealed that the macrophages in the SVF were of the anti-inflammatory phenotype. Western blot analysis showed that the SVF increased Smad2/3 phosphorylation, while Smad2/3 inhibitors decreased phosphorylation. Smad2/3 inhibitors also reduced the expression of various other paracrine factors and decreased chondrocyte pellet size. These findings suggested that the paracrine effect of heterogeneous cells, such as anti-inflammatory macrophages, in the SVF partly supports chondrocyte regeneration through TGF-β-induced Smad2/3 phosphorylation.
Collapse
Affiliation(s)
- Masahiro Fujita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shinya Hayashi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shingo Hashimoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoki Nakano
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Toshihisa Maeda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuichi Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshinori Takashima
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenichi Kikuchi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kensuke Anjiki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kemmei Ikuta
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuma Onoi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shotaro Tachibana
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takehiko Matsushita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hideki Iwaguro
- Department of Orthopaedic Surgery, Sobajima Clinic, Osaka, Japan
| | - Satoshi Sobajima
- Department of Orthopaedic Surgery, Sobajima Clinic, Osaka, Japan
| | - Takafumi Hiranaka
- Department of Orthopaedic Surgery and Joint Surgery Centre, Takatsuki General Hospital, Osaka, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|