1
|
da Silva KN, Marim FM, Rocha GV, Costa-Ferro ZSM, França LSDA, Nonaka CKV, Paredes BD, Rossi EA, Loiola EC, Adanho CSA, Cunha RS, Silva MMAD, Cruz FF, Costa VV, Zanette DL, Rocha CAG, Aguiar RS, Rocco PRM, Souza BSDF. Functional heterogeneity of mesenchymal stem cells and their therapeutic potential in the K18-hACE2 mouse model of SARS-CoV-2 infection. Stem Cell Res Ther 2025; 16:15. [PMID: 39849557 PMCID: PMC11756204 DOI: 10.1186/s13287-024-04086-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/28/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Despite many years of investigation into mesenchymal stem cells (MSCs) and their potential for treating inflammatory conditions such as COVID-19, clinical outcomes remain variable due to factors like donor variability, different tissue sources, and diversity within MSC populations. Variations in MSCs' secretory and proliferation profiles, and their proteomic and transcriptional characteristics significantly influence their therapeutic potency, highlighting the need for enhanced characterization methods to better predict their efficacy. This study aimed to evaluate the biological characteristics of MSCs from different tissue origins, selecting the most promising line for further validation in a K18-hACE2 mouse model of SARS-CoV-2 infection. METHODS We studied nine MSC lines sourced from either bone marrow (hBMMSC), dental pulp (hDPMSC), or umbilical cord tissue (hUCMSC). The cells were assessed for their proliferative capacity, immunophenotype, trilineage differentiation, proteomic profile, and in vitro immunomodulatory potential by co-culture with activated lymphocytes. The most promising MSC line was selected for further experimental validation using the K18-hACE2 mouse model of SARS-CoV-2 infection. RESULTS The analyzed cells met the minimum criteria for defining MSCs, including the expression of surface molecules and differentiation capacity, showing genetic stability and proliferative potential. Proteomic analysis revealed distinct protein profiles that correlate with the tissue origin of MSCs. The immunomodulatory response exhibited variability, lacking a discernible pattern associated with their origin. In co-culture assays with lymphocytes activated with anti-CD3/CD28 beads, all MSC lines demonstrated the ability to inhibit TNF-α, to induce TGF-β and Indoleamine 2,3-dioxygenase (IDO), with varying degrees of inhibition observed for IFN-γ and IL-6, or induction of IL-10 expression. A module of proteins was found to statistically correlate with the potency of IL-6 modulation, leading to the selection of one of the hUCMSCs as the most promising line. Administration of hUCMSC to SARS-CoV-2-infected K18 mice expressing hACE2 was effective in improving lung histology and modulating of a panel of cytokines. CONCLUSIONS Our study assessed MSCs derived from various tissues, uncovering significant variability in their characteristics and immunomodulatory capacities. Particularly, hUCMSCs demonstrated potential in mitigating lung pathology in a SARS-CoV-2 infection model, suggesting their promising therapeutic efficacy.
Collapse
Affiliation(s)
- Kátia Nunes da Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Fernanda Martins Marim
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Gisele Vieira Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | | | | | | | | | - Erik Aranha Rossi
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Erick Correia Loiola
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | | | - Rachel Santana Cunha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Mayck Medeiros Amaral da Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivian Vasconcelos Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Clarissa Araújo Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Renato Santana Aguiar
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno Solano de Freitas Souza
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil.
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil.
| |
Collapse
|
2
|
Wang Y, Thottappillil N, Gomez-Salazar M, Tower RJ, Qin Q, Del Rosario Alvia IC, Xu M, Cherief M, Cheng R, Archer M, Arondekar S, Reddy S, Broderick K, Péault B, James AW. Integrated transcriptomics of human blood vessels defines a spatially controlled niche for early mesenchymal progenitor cells. Dev Cell 2024; 59:2687-2703.e6. [PMID: 39025061 PMCID: PMC11496018 DOI: 10.1016/j.devcel.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/28/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024]
Abstract
Human blood vessel walls show concentric layers, with the outermost tunica adventitia harboring mesenchymal progenitor cells. These progenitor cells maintain vessel homeostasis and provide a robust cell source for cell-based therapies. However, human adventitial stem cell niche has not been studied in detail. Here, using spatial and single-cell transcriptomics, we characterized the phenotype, potential, and microanatomic distribution of human perivascular progenitors. Initially, spatial transcriptomics identified heterogeneity between perivascular layers of arteries and veins and delineated the tunica adventitia into inner and outer layers. From this spatial atlas, we inferred a hierarchy of mesenchymal progenitors dictated by a more primitive cell with a high surface expression of CD201 (PROCR). When isolated from humans and mice, CD201Low expression typified a mesodermal committed subset with higher osteogenesis and less proliferation than CD201High cells, with a downstream effect on canonical Wnt signaling through DACT2. CD201Low cells also displayed high translational potential for bone tissue generation.
Collapse
Affiliation(s)
- Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | | | - Robert J Tower
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Mingxin Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Ray Cheng
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Mary Archer
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Shreya Arondekar
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sashank Reddy
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Kristen Broderick
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Bruno Péault
- Department of Orthopedic Surgery and Orthopedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
3
|
Becker WP, Dáu JBT, Souza WD, Mendez-Otero R, Carias RBV, Jasmin. Incorporation of Superparamagnetic Magnetic–Fluorescent Iron Oxide Nanoparticles Increases Proliferation of Human Mesenchymal Stem Cells. MAGNETOCHEMISTRY 2024; 10:77. [DOI: 10.3390/magnetochemistry10100077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Mesenchymal stem cells (MSCs) have significant therapeutic potential and their use requires in-depth studies to better understand their effects. Labeling cells with superparamagnetic iron oxide nanoparticles allows real-time monitoring of their location, migration, and fate post-transplantation. This study aimed to investigate the efficacy and cytotoxicity of magnetic–fluorescent nanoparticles in human adipose tissue-derived mesenchymal stem cells (hADSCs). The efficacy of Molday ION rhodamine B (MIRB) labeling in hADSCs was evaluated and their biocompatibility was assessed using various techniques and differentiation assays. Prussian blue and fluorescence staining confirmed that 100% of the cells were labeled with MIRB and this labeling persisted for at least 3 days. Transmission electron microscopy revealed the internalization and clustering of the nanoparticles on the outer surface of the cell membrane. The viability assay showed increased cell viability 3 days after nanoparticle exposure. Cell counts were higher in the MIRB-treated group compared to the control group at 3 and 5 days and an increased cell proliferation rate was observed at 3 days post-exposure. Adipogenic, osteogenic, and chondrogenic differentiation was successfully achieved in all groups, with MIRB-treated cells showing an enhanced differentiation rate into adipocytes and osteocytes. MIRB was efficiently internalized by hADSCs but induced changes in cellular behavior due to the increased cell proliferation rate.
Collapse
Affiliation(s)
- Willian Pinheiro Becker
- Núcleo Multidisciplinar de Pesquisa UFRJ-Xerém em Biologia, Universidade Federal do Rio de Janeiro, Duque de Caxias 25240-005, Rio de Janeiro, Brazil
| | - Juliana Barbosa Torreão Dáu
- Centro de Medicina Regenerativa, Faculdade de Medicina de Petrópolis, Petrópolis 25680-120, Rio de Janeiro, Brazil
| | - Wanderson de Souza
- Divisão de Metrologia em Biologia, Instituto Nacional de Metrologia, Qualidade e Tecnologia, Duque de Caxias 25250-020, Rio de Janeiro, Brazil
| | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Duque de Caxias 25240-005, Rio de Janeiro, Brazil
| | - Rosana Bizon Vieira Carias
- Centro de Medicina Regenerativa, Faculdade de Medicina de Petrópolis, Petrópolis 25680-120, Rio de Janeiro, Brazil
| | - Jasmin
- Núcleo Multidisciplinar de Pesquisa UFRJ-Xerém em Biologia, Universidade Federal do Rio de Janeiro, Duque de Caxias 25240-005, Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Reis ALG, Maximino JR, Lage LADPC, Gomes HR, Pereira J, Brofman PRS, Senegaglia AC, Rebelatto CLK, Daga DR, Paiva WS, Chadi G. Proteomic analysis of cerebrospinal fluid of amyotrophic lateral sclerosis patients in the presence of autologous bone marrow derived mesenchymal stem cells. Stem Cell Res Ther 2024; 15:301. [PMID: 39278909 PMCID: PMC11403799 DOI: 10.1186/s13287-024-03820-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 06/27/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly progressive motoneuron degenerative disorder. There are still no drugs capable of slowing disease evolution or improving life quality of ALS patients. Thus, autologous stem cell therapy has emerged as an alternative treatment regime to be investigated in clinical ALS. METHOD Using Proteomics and Protein-Protein Interaction Network analyses combined with bioinformatics, the possible cellular mechanisms and molecular targets related to mesenchymal stem cells (MSCs, 1 × 106 cells/kg, intrathecally in the lumbar region of the spine) were investigated in cerebrospinal fluid (CSF) of ALS patients who received intrathecal infusions of autologous bone marrow-derived MSCs thirty days after cell therapy. Data are available via ProteomeXchange with identifier PXD053129. RESULTS Proteomics revealed 220 deregulated proteins in CSF of ALS subjects treated with MSCs compared to CSF collected from the same patients prior to MSCs infusion. Bioinformatics enriched analyses highlighted events of Extracellular matrix and Cell adhesion molecules as well as related key targets APOA1, APOE, APP, C4A, C5, FGA, FGB, FGG and PLG in the CSF of cell treated ALS subjects. CONCLUSIONS Extracellular matrix and cell adhesion molecules as well as their related highlighted components have emerged as key targets of autologous MSCs in CSF of ALS patients. TRIAL REGISTRATION Clinicaltrial.gov identifier NCT0291768. Registered 28 September 2016.
Collapse
Affiliation(s)
- Ana Luiza Guimarães Reis
- Laboratorio de Neurologia Translacional, Departamento de Neurologia, Hospital das Clinicas, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Jessica Ruivo Maximino
- Laboratorio de Neurologia Translacional, Departamento de Neurologia, Hospital das Clinicas, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | | | - Hélio Rodrigues Gomes
- Departamento de Neurologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Juliana Pereira
- LIM-31, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Paulo Roberto Slud Brofman
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Alexandra Cristina Senegaglia
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Carmen Lúcia Kuniyoshi Rebelatto
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Debora Regina Daga
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Catolica do Parana, Curitiba, PR, 80215-901, Brazil
| | - Wellingson Silva Paiva
- Departamento de Neurologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil
| | - Gerson Chadi
- Laboratorio de Neurologia Translacional, Departamento de Neurologia, Hospital das Clinicas, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil.
- Departamento de Neurologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, SP, 01246-903, Brazil.
| |
Collapse
|
5
|
Tota M, Jonderko L, Witek J, Novickij V, Kulbacka J. Cellular and Molecular Effects of Magnetic Fields. Int J Mol Sci 2024; 25:8973. [PMID: 39201657 PMCID: PMC11354277 DOI: 10.3390/ijms25168973] [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: 07/17/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Recently, magnetic fields (MFs) have received major attention due to their potential therapeutic applications and biological effects. This review provides a comprehensive analysis of the cellular and molecular impacts of MFs, with a focus on both in vitro and in vivo studies. We investigate the mechanisms by which MFs influence cell behavior, including modifications in gene expression, protein synthesis, and cellular signaling pathways. The interaction of MFs with cellular components such as ion channels, membranes, and the cytoskeleton is analyzed, along with their effects on cellular processes like proliferation, differentiation, and apoptosis. Molecular insights are offered into how MFs modulate oxidative stress and inflammatory responses, which are pivotal in various pathological conditions. Furthermore, we explore the therapeutic potential of MFs in regenerative medicine, cancer treatment, and neurodegenerative diseases. By synthesizing current findings, this article aims to elucidate the complex bioeffects of MFs, thereby facilitating their optimized application in medical and biotechnological fields.
Collapse
Affiliation(s)
- Maciej Tota
- Student Research Group № K148, Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Laura Jonderko
- Student Research Group № K148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland; (L.J.); (J.W.)
| | - Julia Witek
- Student Research Group № K148, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wroclaw, Poland; (L.J.); (J.W.)
| | - Vitalij Novickij
- Institute of High Magnetic Fields, Vilnius Gediminas Technical University, LT-03227 Vilnius, Lithuania;
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių 5, LT-08410 Vilnius, Lithuania
| | - Julita Kulbacka
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariškių 5, LT-08410 Vilnius, Lithuania
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 50-367 Wrocław, Poland
| |
Collapse
|
6
|
Motalebzadeh E, Hemati S, Mayvani MA, Ghollasi M. Employing novel biocompatible composite scaffolds with bioglass 58S and poly L-lactic acid for effective bone defect treatment. Mol Biol Rep 2024; 51:838. [PMID: 39042226 DOI: 10.1007/s11033-024-09763-4] [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: 02/23/2024] [Accepted: 06/26/2024] [Indexed: 07/24/2024]
Abstract
BACKGROUND Bioglass materials have gained significant attention in the field of tissue engineering due to their osteoinductive and biocompatible properties that promote bone cell differentiation. In this study, a novel composite scaffold was developed using a sol-gel technique to combine bioglass (BG) 58 S with a poly L-lactic acid (PLLA). METHODS AND RESULTS The physiochemical properties, morphology, and osteoinductive potential of the scaffolds were investigated by X-ray diffraction analysis, scanning electron microscopy, and Fourier-transform infrared spectroscopy. The results showed that the SiO2-CaO-P2O5 system was successfully synthesized by the sol-gel method. The PLLA scaffolds containing BG was found to be osteoinductive and promoted mineralization, as demonstrated by calcium deposition assay, upregulation of alkaline phosphatase enzyme activity, and Alizarin red staining data. CONCLUSIONS These in vitro studies suggest that composite scaffolds incorporating hBMSCs are a promising substitute material to be implemented in bone tissue engineering. The PLLA/BG scaffolds promote osteogenesis and support the differentiation of bone cells, such as osteoblasts, due to their osteoinductive properties.
Collapse
Affiliation(s)
- Erfan Motalebzadeh
- Department of Biology, Basic Science Faculty, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Saideh Hemati
- Department of Cellular and Molecular Biology, Faculty of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohanna Akbarin Mayvani
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Marzieh Ghollasi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| |
Collapse
|
7
|
Castilla-Casadiego DA, Morton LD, Loh DH, Pineda-Hernandez A, Chavda AP, Garcia F, Rosales AM. Peptoid-Cross-Linked Hydrogel Stiffness Modulates Human Mesenchymal Stromal Cell Immunoregulatory Potential in the Presence of Interferon-Gamma. Macromol Biosci 2024; 24:e2400111. [PMID: 38567626 PMCID: PMC11250919 DOI: 10.1002/mabi.202400111] [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: 03/22/2024] [Indexed: 04/04/2024]
Abstract
Human mesenchymal stromal cell (hMSC) manufacturing requires the production of large numbers of therapeutically potent cells. Licensing with soluble cytokines improves hMSC therapeutic potency by enhancing secretion of immunoactive factors but typically decreases proliferative ability. Soft hydrogels, however, have shown promise for boosting immunomodulatory potential, which may compensate for decreased proliferation. Here, hydrogels are cross-linked with peptoids of different secondary structures to generate substrates of various bulk stiffnesses but fixed network connectivity. Secretions of interleukin 6, monocyte chemoattractive protein-1, macrophage colony-stimulating factor, and vascular endothelial growth factor are shown to depend on hydrogel stiffness in the presence of interferon gamma (IFN-γ) supplementation, with soft substrates further improving secretion. The immunological function of these secreted cytokines is then investigated via coculture of hMSCs seeded on hydrogels with primary peripheral blood mononuclear cells (PBMCs) in the presence and absence of IFN-γ. Cocultures with hMSCs seeded on softer hydrogels show decreased PBMC proliferation with IFN-γ. To probe possible signaling pathways, immunofluorescent studies probe the nuclear factor kappa B pathway and demonstrate that IFN-γ supplementation and softer hydrogel mechanics lead to higher activation of this pathway. Overall, these studies may allow for production of more efficacious therapeutic hMSCs in the presence of IFN-γ.
Collapse
Affiliation(s)
| | - Logan D. Morton
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Darren H. Loh
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Aldaly Pineda-Hernandez
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ajay P. Chavda
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Francis Garcia
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Adrianne M. Rosales
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
8
|
Wikarska A, Roszak K, Roszek K. Mesenchymal Stem Cells and Purinergic Signaling in Autism Spectrum Disorder: Bridging the Gap between Cell-Based Strategies and Neuro-Immune Modulation. Biomedicines 2024; 12:1310. [PMID: 38927517 PMCID: PMC11201695 DOI: 10.3390/biomedicines12061310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/26/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of autism spectrum disorder (ASD) is still increasing, which means that this neurodevelopmental lifelong pathology requires special scientific attention and efforts focused on developing novel therapeutic approaches. It has become increasingly evident that neuroinflammation and dysregulation of neuro-immune cross-talk are specific hallmarks of ASD, offering the possibility to treat these disorders by factors modulating neuro-immunological interactions. Mesenchymal stem cell-based therapy has already been postulated as one of the therapeutic approaches for ASD; however, less is known about the molecular mechanisms of stem cell influence. One of the possibilities, although still underestimated, is the paracrine purinergic activity of MSCs, by which stem cells ameliorate inflammatory reactions. Modulation of adenosine signaling may help restore neurotransmitter balance, reduce neuroinflammation, and improve overall brain function in individuals with ASD. In our review article, we present a novel insight into purinergic signaling, including but not limited to the adenosinergic pathway and its role in neuroinflammation and neuro-immune cross-talk modulation. We anticipate that by achieving a greater understanding of the purinergic signaling contribution to ASD and related disorders, novel therapeutic strategies may be devised for patients with autism in the near future.
Collapse
Affiliation(s)
| | | | - Katarzyna Roszek
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Lwowska 1, 87-100 Torun, Poland; (A.W.); (K.R.)
| |
Collapse
|
9
|
Guo T, Pei F, Zhang M, Yamada T, Feng J, Jing J, Ho TV, Chai Y. Vascular architecture regulates mesenchymal stromal cell heterogeneity via P53-PDGF signaling in the mouse incisor. Cell Stem Cell 2024; 31:904-920.e6. [PMID: 38703771 PMCID: PMC11162319 DOI: 10.1016/j.stem.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 02/17/2024] [Accepted: 04/15/2024] [Indexed: 05/06/2024]
Abstract
Mesenchymal stem cells (MSCs) reside in niches to maintain tissue homeostasis and contribute to repair and regeneration. Although the physiological functions of blood and lymphatic vasculature are well studied, their regulation of MSCs as niche components remains largely unknown. Using adult mouse incisors as a model, we uncover the role of Trp53 in regulating vascular composition through THBS2 to maintain mesenchymal tissue homeostasis. Loss of Trp53 in GLI1+ progeny increases arteries and decreases other vessel types. Platelet-derived growth factors from arteries deposit in the MSC region and interact with PDGFRA and PDGFRB. Significantly, PDGFRA+ and PDGFRB+ cells differentially contribute to defined cell lineages in the adult mouse incisor. Collectively, our results highlight Trp53's importance in regulating the vascular niche for MSCs. They also shed light on how different arterial cells provide unique cues to regulate MSC subpopulations and maintain their heterogeneity. Furthermore, they provide mechanistic insight into MSC-vasculature crosstalk.
Collapse
Affiliation(s)
- Tingwei Guo
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Fei Pei
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Mingyi Zhang
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Takahiko Yamada
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Jifan Feng
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Junjun Jing
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Thach-Vu Ho
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
10
|
Ding Z, Greenberg ZF, Serafim MF, Ali S, Jamieson JC, Traktuev DO, March K, He M. Understanding molecular characteristics of extracellular vesicles derived from different types of mesenchymal stem cells for therapeutic translation. EXTRACELLULAR VESICLE 2024; 3:100034. [PMID: 38957857 PMCID: PMC11218754 DOI: 10.1016/j.vesic.2024.100034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Mesenchymal stem cells (MSCs) have been studied for decades as candidates for cellular therapy, and their secretome, including secreted extracellular vesicles (EVs), has been identified to contribute significantly to regenerative and reparative functions. Emerging evidence has suggested that MSC-EVs alone, could be used as therapeutics that emulate the biological function of MSCs. However, just as with MSCs, MSC-EVs have been shown to vary in composition, depending on the tissue source of the MSCs as well as the protocols employed in culturing the MSCs and obtaining the EVs. Therefore, the importance of careful choice of cell sources and culture environments is receiving increasing attention. Many factors contribute to the therapeutic potential of MSC-EVs, including the source tissue, isolation technique, and culturing conditions. This review illustrates the molecular landscape of EVs derived from different types of MSC cells along with culture strategies. A thorough analysis of publicly available omic datasets was performed to advance the precision understanding of MSC-EVs with unique tissue source-dependent molecular characteristics. The tissue-specific protein and miRNA-driven Reactome ontology analysis was used to reveal distinct patterns of top Reactome ontology pathways across adipose, bone marrow, and umbilical MSC-EVs. Moreover, a meta-analysis assisted by an AI technique was used to analyze the published literature, providing insights into the therapeutic translation of MSC-EVs based on their source tissues.
Collapse
Affiliation(s)
- Zuo Ding
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Zachary F. Greenberg
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Maria Fernanda Serafim
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Samantha Ali
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Julia C. Jamieson
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Dmitry O. Traktuev
- UF Center for Regenerative Medicine, Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Keith March
- UF Center for Regenerative Medicine, Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Mei He
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
11
|
Marquez-Curtis LA, Elliott JAW. Mesenchymal stromal cells derived from various tissues: Biological, clinical and cryopreservation aspects: Update from 2015 review. Cryobiology 2024; 115:104856. [PMID: 38340887 DOI: 10.1016/j.cryobiol.2024.104856] [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: 11/28/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
Mesenchymal stromal cells (MSCs) have become one of the most investigated and applied cells for cellular therapy and regenerative medicine. In this update of our review published in 2015, we show that studies continue to abound regarding the characterization of MSCs to distinguish them from other similar cell types, the discovery of new tissue sources of MSCs, and the confirmation of their properties and functions that render them suitable as a therapeutic. Because cryopreservation is widely recognized as the only technology that would enable the on-demand availability of MSCs, here we show that although the traditional method of cryopreserving cells by slow cooling in the presence of 10% dimethyl sulfoxide (Me2SO) continues to be used by many, several novel MSC cryopreservation approaches have emerged. As in our previous review, we conclude from these recent reports that viable and functional MSCs from diverse tissues can be recovered after cryopreservation using a variety of cryoprotectants, freezing protocols, storage temperatures, and periods of storage. We also show that for logistical reasons there are now more studies devoted to the cryopreservation of tissues from which MSCs are derived. A new topic included in this review covers the application in COVID-19 of MSCs arising from their immunomodulatory and antiviral properties. Due to the inherent heterogeneity in MSC populations from different sources there is still no standardized procedure for their isolation, identification, functional characterization, cryopreservation, and route of administration, and not likely to be a "one-size-fits-all" approach in their applications in cell-based therapy and regenerative medicine.
Collapse
Affiliation(s)
- Leah A Marquez-Curtis
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada, T6G 1H9; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada, T6G 1C9
| | - Janet A W Elliott
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada, T6G 1H9; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada, T6G 1C9.
| |
Collapse
|
12
|
Valim Parca A, Godoy Pieri NC, Fantinato Neto P, Fernandes Bressan F, Ambrósio CE, Santos Martins DD. Comparative Analysis of Fluorescent Labeling Techniques for Tracking Canine Amniotic Stem Cells. Tissue Eng Part C Methods 2024; 30:183-192. [PMID: 38411508 DOI: 10.1089/ten.tec.2023.0286] [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: 02/28/2024] Open
Abstract
The utmost aim of regenerative medicine is to promote the regeneration of injured tissues using stem cells. Amniotic mesenchymal stem cells (AmMSCs) have been used in several studies mainly because of their easy isolation from amniotic tissue postpartum and immunomodulatory and angiogenic properties and the low level of rejection. These cells share characteristics of both embryonic/fetal and adult stem cells and are particularly advantageous because they do not trigger tumorigenic activity when injected into immunocompromised animals. The large-scale use of AmMSCs for cellular therapies would greatly benefit from fluorescence labeling studies to validate their tracking in future therapies. This study evaluated the fluorophore positivity, fluorescence intensity, and longevity of canine AmMSCs. For this purpose, canine AmMSCs from the GDTI/USP biobank were submitted to three labeling conditions, two commercial fluorophores [CellTrace CFSE Cell Proliferation kit - CTrace, and CellTracker Green CMFDA - CTracker (CellTracker Green CMFDA, CT, #C2925, Molecular Probes®; Life Technologies)] and green fluorescent protein (GFP) expression after lentiviral transduction, to select the most suitable tracer in terms of adequate persistence and easy handling and analysis that could be used in studies of domestic animals. Fluorescence was detected in all groups; however, the patterns were different. Specifically, CTrace and CTracker fluorescence was detected 6 h after labeling, while GFP was visualized no earlier than 48 h after transduction. Flow cytometry analysis revealed more than 70% of positive cells on day 7 in the CTrace and CTracker groups, while fluorescence decreased significantly to 10% or less on day 20. Variations between repetitions were observed in the GFP group under the present conditions. Our results showed earlier fluorescence detection and more uniform results across repetitions for the commercial fluorophores. In contrast, fluorescence persisted for more extended periods in the GFP group. These results indicate a promising direction for assessing the roles of canine AmMSCs in regenerative medicine without genomic integration.
Collapse
Affiliation(s)
- Andressa Valim Parca
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Naira Caroline Godoy Pieri
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Paulo Fantinato Neto
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Fabiana Fernandes Bressan
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Carlos Eduardo Ambrósio
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Daniele Dos Santos Martins
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
| |
Collapse
|
13
|
Alshamsi MAH, Mosa KA, Khan AA, Mousa M, Ali MA, Soliman SSM, Semreen MH. Biosynthesized Silver Nanoparticles from Cyperus conglomeratus Root Extract Inhibit Osteogenic Differentiation of Immortalized Mesenchymal Stromal Cells. Curr Pharm Biotechnol 2024; 25:1333-1347. [PMID: 37612859 DOI: 10.2174/1389201024666230823094412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/23/2023] [Accepted: 07/20/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND Silver nanoparticles (AgNPs) are a focus of huge interest in biological research, including stem cell research. AgNPs synthesized using Cyperus conglomeratus root extract have been previously reported but their effects on mesenchymal stromal cells have yet to be investigated. OBJECTIVES The aim of this study is to investigate the effects of C. conglomeratus-derived AgNPs on adipogenesis and osteogenesis of mesenchymal stromal cells. METHODS AgNPs were synthesized using C. conglomeratus root extract, and the phytochemicals involved in AgNPs synthesis were analyzed using gas chromatography-mass spectrometry (GCMS). The cytotoxicity of the AgNPs was tested on telomerase-transformed immortalized human bone marrow-derived MSCs-hTERT (iMSC3) and human osteosarcoma cell line (MG-63) using MTT and apoptosis assays. The uptake of AgNPs by both cells was confirmed using inductively coupled plasma-optical emission spectrometry (ICP-OES). Furthermore, the effect of AgNPs on iMSC3 adipogenesis and osteogenesis was analyzed using stain quantification and reverse transcription- quantitative polymerase chain reaction (RT-qPCR). RESULTS The phytochemicals predominately identified in both the AgNPs and C. conglomeratus root extract were carbohydrates. The AgNP concentrations tested using MTT and apoptosis assays (0.5-64 µg/ml and 1,4 and 32 µg/ml, respectively) showed no significant cytotoxicity on iMSC3 and MG-63. The AgNPs were internalized in a concentration-dependent manner in both cell types. Additionally, the AgNPs exhibited a significant negative effect on osteogenesis but not on adipogenesis. CONCLUSION C. conglomeratus-derived AgNPs had an impact on the differentiation capacity of iMSC3. Our results indicated that C. conglomeratus AgNPs and the associated phytochemicals could exhibit potential medical applications.
Collapse
Affiliation(s)
- Mohamed A H Alshamsi
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Kareem A Mosa
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Biotechnology, Faculty of Agriculture, Al-Azhar University, Cairo, Egypt
| | - Amir Ali Khan
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Muath Mousa
- Research Institute of Science and Engineering (RISE), University of Sharjah, Sharjah, United Arab Emirates
| | - Muna A Ali
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Sameh S M Soliman
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Mohammad H Semreen
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
14
|
Barchiki F, Fracaro L, Dominguez AC, Senegaglia AC, Vaz IM, Soares P, de Moura SAB, Brofman PRS. Biocompatibility of ABS and PLA Polymers with Dental Pulp Stem Cells Enhance Their Potential Biomedical Applications. Polymers (Basel) 2023; 15:4629. [PMID: 38139880 PMCID: PMC10747830 DOI: 10.3390/polym15244629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Polylactic Acid (PLA) and Acrylonitrile-Butadiene-Styrene (ABS) are commonly used polymers in 3D printing for biomedical applications. Dental Pulp Stem Cells (DPSCs) are an accessible and proliferative source of stem cells with significant differentiation potential. Limited knowledge exists regarding the biocompatibility and genetic safety of ABS and PLA when in contact with DPSCs. This study aimed to investigate the impact of PLA and ABS on the adhesion, proliferation, osteogenic differentiation, genetic stability, proteomics, and immunophenotypic profile of DPSCs. A total of three groups, 1- DPSC-control, 2- DPSC+ABS, and 3- DPSC+PLA, were used in in vitro experiments to evaluate cell morphology, proliferation, differentiation capabilities, genetic stability, proteomics (secretome), and immunophenotypic profiles regarding the interaction between DPSCs and polymers. Both ABS and PLA supported the adhesion and proliferation of DPSCs without exhibiting significant cytotoxic effects and maintaining the capacity for osteogenic differentiation. Genetic stability, proteomics, and immunophenotypic profiles were unaltered in DPSCs post-contact with these polymers, highlighting their biosafety. Our findings suggest that ABS and PLA are biocompatible with DPSCs and demonstrate potential in dental or orthopedic applications; the choice of the polymer will depend on the properties required in treatment. These promising results stimulate further studies to explore the potential therapeutic applications in vivo using prototyped polymers in personalized medicine.
Collapse
Affiliation(s)
- Fabiane Barchiki
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, Brazil; (L.F.); (A.C.S.); (I.M.V.); (P.R.S.B.)
- INCT—REGENERA National Institute of Science and Technology in Regenerative Medicine, Rio de Janeiro 21941-902, Brazil
| | - Letícia Fracaro
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, Brazil; (L.F.); (A.C.S.); (I.M.V.); (P.R.S.B.)
- INCT—REGENERA National Institute of Science and Technology in Regenerative Medicine, Rio de Janeiro 21941-902, Brazil
| | - Alejandro Correa Dominguez
- Laboratory of Basic Biology of Stem Cells, Carlos Chagas Institute, Fiocruz-PR, Curitiba 81350-010, Brazil;
| | - Alexandra Cristina Senegaglia
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, Brazil; (L.F.); (A.C.S.); (I.M.V.); (P.R.S.B.)
- INCT—REGENERA National Institute of Science and Technology in Regenerative Medicine, Rio de Janeiro 21941-902, Brazil
| | - Isadora May Vaz
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, Brazil; (L.F.); (A.C.S.); (I.M.V.); (P.R.S.B.)
- INCT—REGENERA National Institute of Science and Technology in Regenerative Medicine, Rio de Janeiro 21941-902, Brazil
| | - Paulo Soares
- LaBES—Laboratory of Biomaterials and Surface Engineering, Polytechnic School, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, Brazil;
| | - Sérgio Adriane Bezerra de Moura
- Departament of Morphology, Campus Universitário Lagoa Nova, Universidade Federal do Rio Grande do Norte (UFRN), Natal 59072-970, Brazil;
| | - Paulo Roberto Slud Brofman
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, Brazil; (L.F.); (A.C.S.); (I.M.V.); (P.R.S.B.)
- INCT—REGENERA National Institute of Science and Technology in Regenerative Medicine, Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
15
|
Zheng M, Zhang H, Wu H, Xie J, Chen Q, Jiang Y, Zhao D. Assessing the role of combination of stem cell and light-based treatments on skin wound repair: A meta-analysis. Int Wound J 2023; 20:4272-4280. [PMID: 37525509 PMCID: PMC10681544 DOI: 10.1111/iwj.14329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 08/02/2023] Open
Abstract
The meta-analysis aims to evaluate and compare the impact of the combination of stem cells (SCs) and light-based treatments (LBTs) on skin wound (SW) repair. Examinations comparing SCs to LBT with SCs for SW repair was among the meta-analysis from various languages that met the inclusion criteria. Using continuous random-effect models, the results of these investigations were examined, and the mean difference (MD) with 95% confidence intervals was computed (CIs). Seven examinations from 2012 to 2022 were recruited for the current analysis including 106 animals with SWs. Photobiomodulation therapy (PBT) plus SCs had a significantly higher wound closure rate (WCR) (MD, 9.08; 95% CI, 5.55-12.61, p < 0.001) compared to SCs in animals with SWs. However, no significant difference was found between PBT plus SCs and SCs on wound tensile strength (WTS) (MD, 2.01; 95% CI, -0.42 to 4.44, p = 0.10) in animals with SWs. The examined data revealed that PBT plus SCs had a significantly higher WCR, however, no significant difference was found in WTS compared to SCs in animals with SWs. Nevertheless, caution should be exercised while interacting with its values since all the chosen examinations were found with a low sample size and a low number of examinations were found for the comparisons studied for the meta-analysis.
Collapse
Affiliation(s)
- Mingjing Zheng
- Department of DermatologyWenzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese MedicineWenzhouChina
| | - Huihe Zhang
- Department of NeurologyWenzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese MedicineWenzhouChina
| | - Huizhen Wu
- Department of Dermatologythe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Jiayi Xie
- Graduate CollegeZhejiang Chinese Medical UniversityHangzhouChina
| | - Qiong Chen
- Department of SurgeryWenzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese MedicineWenzhouChina
| | - Yue Jiang
- Department of Acupuncturethe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Dongrui Zhao
- Department of DermatologyWenzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese MedicineWenzhouChina
| |
Collapse
|
16
|
Guarnier LP, Moro LG, Lívero FADR, de Faria CA, Azevedo MF, Roma BP, Albuquerque ER, Malagutti-Ferreira MJ, Rodrigues AGD, da Silva AA, Sekiya EJ, Ribeiro-Paes JT. Regenerative and translational medicine in COPD: hype and hope. Eur Respir Rev 2023; 32:220223. [PMID: 37495247 PMCID: PMC10369169 DOI: 10.1183/16000617.0223-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 05/23/2023] [Indexed: 07/28/2023] Open
Abstract
COPD is a common, preventable and usually progressive disease associated with an enhanced chronic inflammatory response in the airways and lung, generally caused by exposure to noxious particles and gases. It is a treatable disease characterised by persistent respiratory symptoms and airflow limitation due to abnormalities in the airways and/or alveoli. COPD is currently the third leading cause of death worldwide, representing a serious public health problem and a high social and economic burden. Despite significant advances, effective clinical treatments have not yet been achieved. In this scenario, cell-based therapies have emerged as potentially promising therapeutic approaches. However, there are only a few published studies of cell-based therapies in human patients with COPD and a small number of ongoing clinical trials registered on clinicaltrials.gov Despite the advances and interesting results, numerous doubts and questions remain about efficacy, mechanisms of action, culture conditions, doses, timing, route of administration and conditions related to homing and engraftment of the infused cells. This article presents the state of the art of cell-based therapy in COPD. Clinical trials that have already been completed and with published results are discussed in detail. We also discuss the questions that remain unanswered about cell-based regenerative and translational medicine for COPD.
Collapse
Affiliation(s)
- Lucas Pires Guarnier
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
- Laboratory of Genetics and Cell Therapy - GenTe Cel, Department of Biotechnology, São Paulo State University (UNESP), Assis, Brazil
| | - Lincoln Gozzi Moro
- Laboratory of Genetics and Cell Therapy - GenTe Cel, Department of Biotechnology, São Paulo State University (UNESP), Assis, Brazil
- Biomedical Sciences Institute, Butantan Institute, Technological Research Institute, University of São Paulo (USP), São Paulo, Brazil
| | | | | | - Mauricio Fogaça Azevedo
- Laboratory of Genetics and Cell Therapy - GenTe Cel, Department of Biotechnology, São Paulo State University (UNESP), Assis, Brazil
| | - Beatriz Pizoni Roma
- Laboratory of Genetics and Cell Therapy - GenTe Cel, Department of Biotechnology, São Paulo State University (UNESP), Assis, Brazil
| | | | - Maria José Malagutti-Ferreira
- Laboratory of Genetics and Cell Therapy - GenTe Cel, Department of Biotechnology, São Paulo State University (UNESP), Assis, Brazil
| | | | - Adelson Alves da Silva
- São Lucas Research and Education Institute (IEP - São Lucas), TechLife, São Paulo, Brazil
| | - Eliseo Joji Sekiya
- São Lucas Research and Education Institute (IEP - São Lucas), TechLife, São Paulo, Brazil
| | - João Tadeu Ribeiro-Paes
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
- Laboratory of Genetics and Cell Therapy - GenTe Cel, Department of Biotechnology, São Paulo State University (UNESP), Assis, Brazil
| |
Collapse
|
17
|
James AW, Thottappillil N, Péault B, Zhang X. Editorial: Chondrogenic potentials, protocols and mechanisms of mesenchymal progenitor cells. Front Cell Dev Biol 2023; 11:1289438. [PMID: 37786809 PMCID: PMC10541955 DOI: 10.3389/fcell.2023.1289438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 10/04/2023] Open
Affiliation(s)
- Aaron W. James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | | | - Bruno Péault
- Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, CA, United States
| | - Xinli Zhang
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
18
|
Zahra D, Shokat Z, Ahmad A, Javaid A, Khurshid M, Ashfaq UA, Nashwan AJ. Exploring the recent developments of alginate silk fibroin material for hydrogel wound dressing: A review. Int J Biol Macromol 2023; 248:125989. [PMID: 37499726 DOI: 10.1016/j.ijbiomac.2023.125989] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Hydrogels, a type of polymeric material capable of retaining water within a three-dimensional network, have demonstrated their potential in wound healing, surpassing traditional wound dressings. These hydrogels possess remarkable mechanical, chemical, and biological properties, making them suitable scaffolds for tissue regeneration. This article aims to emphasize the advantages of alginate, silk fibroin, and hydrogel-based wound dressings, specifically highlighting their crucial functions that accelerate the healing process of skin wounds. Noteworthy functions include self-healing ability, water solubility, anti-inflammatory properties, adhesion, antimicrobial properties, drug delivery, conductivity, and responsiveness to stimuli. Moreover, recent advancements in hydrogel technology have resulted in the development of wound dressings with enhanced features for monitoring wound progression, further augmenting their effectiveness. This review emphasizes the utilization of hydrogel membranes for treating excisional and incisional wounds, while exploring recent breakthroughs in hydrogel wound dressings, including nanoparticle composite hydrogels, stem cell hydrogel composites, and curcumin-hydrogel composites. Additionally, the review focuses on diverse synthesis procedures, designs, and potential applications of hydrogels in wound healing dressings.
Collapse
Affiliation(s)
- Duaa Zahra
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Pakistan
| | - Zeeshan Shokat
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Pakistan
| | - Azka Ahmad
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Pakistan
| | - Anam Javaid
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Pakistan
| | - Mohsin Khurshid
- Institute of Microbiology, Government College University Faisalabad, Pakistan
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Pakistan.
| | | |
Collapse
|
19
|
Couto PS, Stibbs DJ, Rotondi MC, Takeuchi Y, Rafiq QA. Scalable manufacturing of gene-modified human mesenchymal stromal cells with microcarriers in spinner flasks. Appl Microbiol Biotechnol 2023; 107:5669-5685. [PMID: 37470820 PMCID: PMC10439856 DOI: 10.1007/s00253-023-12634-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/06/2023] [Accepted: 06/11/2023] [Indexed: 07/21/2023]
Abstract
Due to their immunomodulatory properties and in vitro differentiation ability, human mesenchymal stromal cells (hMSCs) have been investigated in more than 1000 clinical trials over the last decade. Multiple studies that have explored the development of gene-modified hMSC-based products are now reaching early stages of clinical trial programmes. From an engineering perspective, the challenge lies in developing manufacturing methods capable of producing sufficient doses of ex vivo gene-modified hMSCs for clinical applications. This work demonstrates, for the first time, a scalable manufacturing process using a microcarrier-bioreactor system for the expansion of gene-modified hMSCs. Upon isolation, umbilical cord tissue mesenchymal stromal cells (UCT-hMSCs) were transduced using a lentiviral vector (LV) with green fluorescent protein (GFP) or vascular endothelial growth factor (VEGF) transgenes. The cells were then seeded in 100 mL spinner flasks using Spherecol microcarriers and expanded for seven days. After six days in culture, both non-transduced and transduced cell populations attained comparable maximum cell concentrations (≈1.8 × 105 cell/mL). Analysis of the culture supernatant identified that glucose was fully depleted after day five across the cell populations. Lactate concentrations observed throughout the culture reached a maximum of 7.5 mM on day seven. Immunophenotype analysis revealed that the transduction followed by an expansion step was not responsible for the downregulation of the cell surface receptors used to identify hMSCs. The levels of CD73, CD90, and CD105 expressing cells were above 90% for the non-transduced and transduced cells. In addition, the expression of negative markers (CD11b, CD19, CD34, CD45, and HLA-DR) was also shown to be below 5%, which is aligned with the criteria established for hMSCs by the International Society for Cell and Gene Therapy (ISCT). This work provides a foundation for the scalable manufacturing of gene-modified hMSCs which will overcome a significant translational and commercial bottleneck. KEY POINTS: • hMSCs were successfully transduced by lentiviral vectors carrying two different transgenes: GFP and VEGF • Transduced hMSCs were successfully expanded on microcarriers using spinner flasks during a period of 7 days • The genetic modification step did not cause any detrimental impact on the hMSC immunophenotype characteristics.
Collapse
Affiliation(s)
- Pedro Silva Couto
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT UK
| | - Dale J. Stibbs
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT UK
| | - Marco C. Rotondi
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT UK
| | - Yasuhiro Takeuchi
- Division of Infection and Immunity, University College London, Gower Street, London, WC1E 6BT UK
- Biotherapeutics and Advanced Therapies, Scientific Research and Innovation, Medicines, and Healthcare Products Regulatory Agency, South Mimms, EN6 3QG UK
| | - Qasim A. Rafiq
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT UK
| |
Collapse
|
20
|
Iqbal F, Johnston A, Wyse B, Rabani R, Mander P, Hoseini B, Wu J, Li RK, Gauthier-Fisher A, Szaraz P, Librach C. Combination human umbilical cord perivascular and endothelial colony forming cell therapy for ischemic cardiac injury. NPJ Regen Med 2023; 8:45. [PMID: 37626067 PMCID: PMC10457300 DOI: 10.1038/s41536-023-00321-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Cell-based therapeutics are promising interventions to repair ischemic cardiac tissue. However, no single cell type has yet been found to be both specialized and versatile enough to heal the heart. The synergistic effects of two regenerative cell types including endothelial colony forming cells (ECFC) and first-trimester human umbilical cord perivascular cells (FTM HUCPVC) with endothelial cell and pericyte properties respectively, on angiogenic and regenerative properties were tested in a rat model of myocardial infarction (MI), in vitro tube formation and Matrigel plug assay. The combination of FTM HUCPVCs and ECFCs synergistically reduced fibrosis and cardiomyocyte apoptosis, while promoting favorable cardiac remodeling and contractility. These effects were in part mediated by ANGPT2, PDGF-β, and VEGF-C. PDGF-β signaling-dependent synergistic effects on angiogenesis were also observed in vitro and in vivo. FTM HUCPVCs and ECFCs represent a cell combination therapy for promoting and sustaining vascularization following ischemic cardiac injury.
Collapse
Affiliation(s)
- Farwah Iqbal
- Create Fertility Centre, Toronto, ON, Canada
- Virginia Tech Carillion School of Medicine, Roanoke, VA, USA
| | | | | | | | | | | | - Jun Wu
- Toronto General Research Institute (TGRI), University Health Network (UHN), Toronto, ON, Canada
| | - Ren-Ke Li
- Toronto General Research Institute (TGRI), University Health Network (UHN), Toronto, ON, Canada
| | | | | | - Clifford Librach
- Create Fertility Centre, Toronto, ON, Canada.
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada.
- Institute of Medical Sciences, Department of Physiology, University of Toronto, Toronto, ON, Canada.
- Department of Obstetrics and Gynecology, Women's College Hospital, Toronto, ON, Canada.
| |
Collapse
|
21
|
Malagutti-Ferreira MJ, Crispim BA, Barufatti A, Cardoso SS, Guarnier LP, Rodríguez FF, Soares MR, Antunes RNS, Ribeiro-Paes JT. Genomic instability in long-term culture of human adipose-derived mesenchymal stromal cells. Braz J Med Biol Res 2023; 56:e12713. [PMID: 37493771 PMCID: PMC10361644 DOI: 10.1590/1414-431x2023e12713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 06/21/2023] [Indexed: 07/27/2023] Open
Abstract
Mesenchymal stromal/stem cells stem (MSC) have been widely studied due to their great potential for application in tissue engineering and regenerative and translational medicine. In MSC-based therapy for human diseases, cell proliferation is required to obtain a large and adequate number of cells to ensure therapeutic efficacy. During in vitro culture, cells are under an artificial environment and manipulative stress that can affect genetic stability. Several regulatory agencies have established guidelines to ensure greater safety in cell-based regenerative and translational medicine, but there is no specific definition about the maximum number of passages that ensure the lowest possible risk in MSC-based regenerative medicine. In this context, the aim of this study was to analyze DNA damage and chromosome alterations in adipose-derived mesenchymal stromal cells (ADMSC) until the eleventh passage and to provide additional subsidies to regulatory agencies related to number of passages in these cells. Thus, two methods in genetic toxicology were adopted: comet assay and micronucleus test. The comet assay results showed an increase in DNA damage from the fifth passage onwards. The micronucleus test showed a statistically significant increase of micronucleus from the seventh passage onwards, indicating a possible mutagenic effect associated with the increase in the number of passages. Based on these results, it is important to emphasize the need to assess genetic toxicology and inclusion of new guidelines by regulatory agencies to guarantee the safety of MSC-based therapies for human diseases.
Collapse
Affiliation(s)
- M J Malagutti-Ferreira
- Departamento de Biotecnologia, Faculdade de Ciências e Letras, Universidade Estadual Paulista, Assis, SP, Brasil
| | - B A Crispim
- Faculdade de Ciências Biológicas e Ambientais, Universidade Federal da Grande Dourados, Dourados, MS, Brasil
| | - A Barufatti
- Faculdade de Ciências Biológicas e Ambientais, Universidade Federal da Grande Dourados, Dourados, MS, Brasil
| | - S S Cardoso
- Faculdade de Ciências Biológicas e Ambientais, Universidade Federal da Grande Dourados, Dourados, MS, Brasil
| | - L P Guarnier
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - F F Rodríguez
- Departamento de Biotecnologia, Faculdade de Ciências e Letras, Universidade Estadual Paulista, Assis, SP, Brasil
| | - M R Soares
- Departamento de Ginecologia e Obstetrícia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - R N S Antunes
- Faculdade de Medicina de Marília (FAMEMA), Hemocentro de Marília, Laboratório de Citometria de Fluxo, Marília, SP, Brasil
| | - J T Ribeiro-Paes
- Departamento de Biotecnologia, Faculdade de Ciências e Letras, Universidade Estadual Paulista, Assis, SP, Brasil
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| |
Collapse
|
22
|
Shimizu Y, Ntege EH, Azuma C, Uehara F, Toma T, Higa K, Yabiku H, Matsuura N, Inoue Y, Sunami H. Management of Rheumatoid Arthritis: Possibilities and Challenges of Mesenchymal Stromal/Stem Cell-Based Therapies. Cells 2023; 12:1905. [PMID: 37508569 PMCID: PMC10378234 DOI: 10.3390/cells12141905] [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: 05/06/2023] [Revised: 06/28/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Rheumatoid arthritis (RA) is a highly prevalent, chronic, and progressive autoimmune disorder primarily affecting joints and muscles. The associated inflammation, pain, and motor restriction negatively impact patient quality of life (QOL) and can even contribute to premature mortality. Further, conventional treatments such as antiinflammatory drugs are only symptomatic. Substantial progress has been made on elucidating the etiopathology of overt RA, in particular the contributions of innate and adaptive immune system dysfunction to chronic inflammation. Although the precise mechanisms underlying onset and progression remain elusive, the discovery of new drug targets, early diagnosis, and new targeted treatments have greatly improved the prognosis and QOL of patients with RA. However, a sizable proportion of patients develop severe adverse effects, exhibit poor responses, or cannot tolerate long-term use of these drugs, necessitating more effective and safer therapeutic alternatives. Mounting preclinical and clinical evidence suggests that the transplantation of multipotent adult stem cells such as mesenchymal stromal/stem cells is a safe and effective treatment strategy for controlling chronic inflammation and promoting tissue regeneration in patients with intractable diseases, including RA. This review describes the current status of MSC-based therapies for RA as well as the opportunities and challenges to broader clinical application.
Collapse
Affiliation(s)
- Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Edward Hosea Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Chinatsu Azuma
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Fuminari Uehara
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Takashi Toma
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Kotaro Higa
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Hiroki Yabiku
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Naoki Matsuura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Yoshikazu Inoue
- Department of Plastic and Reconstructive Surgery, School of Medicine, Fujita Health University, Toyoake 470-1192, Japan
| | - Hiroshi Sunami
- Center for Advanced Medical Research, School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| |
Collapse
|
23
|
Gomez-Salazar MA, Wang Y, Thottappillil N, Hardy RW, Alexandre M, Höller F, Martin N, Gonzalez-Galofre ZN, Stefancova D, Medici D, James AW, Péault B. Aldehyde Dehydrogenase, a Marker of Normal and Malignant Stem Cells, Typifies Mesenchymal Progenitors in Perivascular Niches. Stem Cells Transl Med 2023; 12:474-484. [PMID: 37261440 PMCID: PMC10651226 DOI: 10.1093/stcltm/szad024] [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: 06/22/2022] [Accepted: 04/07/2023] [Indexed: 06/02/2023] Open
Abstract
Innate mesenchymal stem cells exhibiting multilineage differentiation and tissue (re)generative-or pathogenic-properties reside in perivascular niches. Subsets of these progenitors are committed to either osteo-, adipo-, or fibrogenesis, suggesting the existence of a developmental organization in blood vessel walls. We evaluated herein the activity of aldehyde dehydrogenase, a family of enzymes catalyzing the oxidation of aldehydes into carboxylic acids and a reported biomarker of normal and malignant stem cells, within human adipose tissue perivascular areas. A progression of ALDHLow to ALDHHigh CD34+ cells was identified in the tunica adventitia. Mesenchymal stem cell potential was confined to ALDHHigh cells, as assessed by proliferation and multilineage differentiation in vitro of cells sorted by flow cytometry with a fluorescent ALDH substrate. RNA sequencing confirmed and validated that ALDHHigh cells have a progenitor cell phenotype and provided evidence that the main isoform in this fraction is ALDH1A1, which was confirmed by immunohistochemistry. This demonstrates that ALDH activity, which marks hematopoietic progenitors and stem cells in diverse malignant tumors, also typifies native, blood vessel resident mesenchymal stem cells.
Collapse
Affiliation(s)
- Mario A Gomez-Salazar
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
- Department of Pathology, Johns Hopkins University, Baltimore, MB, USA
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MB, USA
| | | | - Reef W Hardy
- Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Manon Alexandre
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
- Polytech Marseille, Aix Marseille University, Marseille, France
| | - Fabian Höller
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Niall Martin
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Zaniah N Gonzalez-Galofre
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Dorota Stefancova
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Daniele Medici
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MB, USA
| | - Bruno Péault
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
- Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
24
|
Afsartala Z, Hadjighassem M, Shirian S, Ebrahimi-Barough S, Gholami L, Hussain MF, Yaghoobi M, Ai J. Advances in Management of Spinal Cord Injury Using Stem Cell-derived Extracellular Vesicles: A Review Study. Basic Clin Neurosci 2023; 14:443-451. [PMID: 38050575 PMCID: PMC10693808 DOI: 10.32598/bcn.2022.3430.2] [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: 06/27/2021] [Revised: 09/03/2021] [Accepted: 12/04/2021] [Indexed: 12/06/2023] Open
Abstract
Introduction Spinal cord injury (SCI) is characterized by serious both motor and sensory disability of the limbs below the injured segment. It is the most traumatic disorder among central nervous system (CNS) conditions which not only leads to psychological and physical harm to patients but also results in a dramatic loss in the life quality. Many efforts have been developed to find a therapeutic approach for SCI; however, an effective treatment has not yet been found. The lack of effective treatment approach and rehabilitation of SCI underscores the need to identify novel approaches. Tissue engineering associated with stem cells has been recently introduced as an effective treatment approaches for traumatic SCI. Although, low survival rates, immune rejection, cell dedifferentiation, and tumorigenicity have been addressed for tissue engineering. Regenerative medicine is an interdisciplinary field developing and applying tissue engineering, stem cell (SC) therapy, and SC-derived extracellular vesicle therapy that aims to provide reliable and safe ways to replace injured tissues and organs. The application of mesenchymal stem cells-derived extracellular vesicles (MSC-EVs) has recently attracted attention to improve central nervous system dysfunction such as SCI, mainly by promoting neurogenesis and angiogenesis. Methods In this review article the latest information of SCI improvement using stem cell-derived extracellular vesicles published data in the Web of Science, Scopus, Science Direct and Pub Med databases were collected. Results The data collected show that MSC-EVs, including exosomes, alone or in combination with scaffolds can can regenerate the injured nerve in SCI. Conclusion This study summarizes the efficacy of MSC-EVs, including exosomes, alone or in combination with scaffolds in the treatment of SCI and then discusses the therapeutic outcomes observed in SCI experimental models following treatment with MSC-EVs alone or loaded on scaffolds in particular collagen-based scaffolds. Highlights The pathological process of SCI being very complex.A complete effective strategy has yet to be found for treatment of SCI in human.Exosomes derived-stem cells alone have great potential for the treatment of SCI.Various biocompatible scaffolds are good drug carriers for SCI treatment.Various biocompatible scaffolds are good carriers for exosomes. Plain Language Summary Human with spinal cord injury (SCI) show serious motor and sensory disability of the limbs. Since there is no an effective treatment for SCI, researchers are trying to develop and find a new therapeutic approach for SCI. CNS tissue engineering with various stem cells sources as well as their derived extracellular vesicle has been extensively attracted for providing reliable and safe approach for SCI treatment. Extracellular vesicles are lipid bilayer membrane-enclosed organelles containing various biomolecules involved in a variety of complex intercellular communication systems. They are released from all cell types into their surrounding environment and are important vehicles for paracrine The application of stem cells-derived extracellular vesicles (MSC-EVs) has recently attracted attention to improve central nervous system dysfunction such as SCI, mainly by promoting neurogenesis and angiogenesis.
Collapse
Affiliation(s)
- Zohreh Afsartala
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoudreza Hadjighassem
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
- Dr. Daneshbod Labratorary, Shiraz Molecular Pathology Research Center, Shiraz, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Gholami
- Department of Periodontics, Dental Research Center, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Fahad Hussain
- Department of Biotechnology, Institute of Biochemical Procedures and Analyzes (IBVA), Technische Hochshule Mittelhessen, Wiesenstr, Germany
| | - Mina Yaghoobi
- Department of Cell Therapy, Medwin Medical Center, Dubai, United Arab Emirates
| | - Jafar Ai
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Park JH, Choi Y, Shin JM, Yang HW, Jeong SH, Park IH. Ultrasound cavitation: a reliable non-enzymatic method for adipose-derived mesenchymal stem cell (ADSC) isolation. Stem Cell Res Ther 2023; 14:153. [PMID: 37280695 DOI: 10.1186/s13287-023-03383-8] [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: 09/27/2022] [Accepted: 05/22/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Adipose tissue is known to serve as an abundant and readily accessible source of adipose-derived stem cells (ADSCs) as an alternative to bone marrow. Collagenase is one of the most widely used methods for the isolation of ADSCs from adipose tissue, but it takes a long time, and there are also debates about safety. We propose an ultrasonic cavitation-treated method that can significantly reduce time and avoid the problem of using xenogeneic enzymes in ADSCs isolation. METHODS ADSCs were isolated from adipose tissue using the enzyme treatment method and the ultrasonic cavitation treatment method. Cell proliferation was measured using cell viability assay. The expression levels of the surface markers of ADSCs were estimated by real-time PCR. After, ADSCs were cultured in chondrogenic, osteogenic, or adipogenic differentiation medium; the differentiation potential of ADCSs was analyzed by Alcian blue, Alizarin Red S, Oil Red O, and real-time PCR. RESULTS The cells treated with collagenase and ultrasound had similar cell yields and proliferation after isolation. The difference in the expression of surface markers of ADSCs was not statistically significant. ADSCs showed differentiation potential into adipocytes, osteocytes, and chondrocytes, and there was no difference between the enzyme treatment method and the ultrasonic cavitation treatment method. The yield of the ADSC increased in time- and intensity dependently. CONCLUSIONS Ultrasound certainly serves as a promising method in advancing ADSC isolation technology.
Collapse
Affiliation(s)
- Joo-Hoo Park
- Upper Airway Chronic Inflammatory Diseases Laboratory, College of Medicine, Korea University, Seoul, Korea
- Medical Devices Usability Test Center, College of Medicine, Korea University, Seoul, Korea
| | - Yujin Choi
- Upper Airway Chronic Inflammatory Diseases Laboratory, College of Medicine, Korea University, Seoul, Korea
| | - Jae-Min Shin
- Upper Airway Chronic Inflammatory Diseases Laboratory, College of Medicine, Korea University, Seoul, Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Guro Hospital, Korea University College of Medicine, 80 Guro-Dong, Guro-Gu, Seoul, 152-703, Korea
- Medical Devices Usability Test Center, College of Medicine, Korea University, Seoul, Korea
| | - Hyun-Woo Yang
- Upper Airway Chronic Inflammatory Diseases Laboratory, College of Medicine, Korea University, Seoul, Korea
- Medical Devices Usability Test Center, College of Medicine, Korea University, Seoul, Korea
| | - Seong-Ho Jeong
- Medical Devices Usability Test Center, College of Medicine, Korea University, Seoul, Korea.
- Department of Plastic Surgery, Guro Hospital, Korea University College of Medicine, 80 Guro-Dong, Guro-Gu, Seoul, 152-703, Korea.
| | - Il-Ho Park
- Upper Airway Chronic Inflammatory Diseases Laboratory, College of Medicine, Korea University, Seoul, Korea.
- Department of Otorhinolaryngology-Head and Neck Surgery, Guro Hospital, Korea University College of Medicine, 80 Guro-Dong, Guro-Gu, Seoul, 152-703, Korea.
- Medical Devices Usability Test Center, College of Medicine, Korea University, Seoul, Korea.
| |
Collapse
|
26
|
Todorova VB, Baxan N, Delahaye M, Harding SE, Rankin SM. Drug-based mobilisation of mesenchymal stem/stromal cells improves cardiac function post myocardial infarction. Dis Model Mech 2023; 16:dmm049630. [PMID: 36263604 PMCID: PMC10655717 DOI: 10.1242/dmm.049630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/14/2022] [Indexed: 11/20/2022] Open
Abstract
There is an unmet need for treatments that prevent the progressive cardiac dysfunction following myocardial infarction. Mesenchymal stem/stromal cells (MSCs) are under investigation for cardiac repair; however, culture expansion prior to transplantation is hindering their homing and reparative abilities. Pharmacological mobilisation could be an alternative to MSC transplantation. Here, we report that endogenous MSCs mobilise into the circulation at day 5 post myocardial infarction in male Lewis rats. This mobilisation can be significantly increased by using a combination of the FDA-approved drugs mirabegron (β3-adrenoceptor agonist) and AMD3100 (CXCR4 antagonist). Blinded cardiac magnetic resonance imaging analysis showed the treated group to have increased left ventricular ejection fraction and decreased end systolic volume at 5 weeks post myocardial infarction. The mobilised group had a significant decrease in plasma IL-6 and TNF-α levels, a decrease in interstitial fibrosis, and an increase in the border zone blood vessel density. Conditioned medium from blood-derived MSCs supported angiogenesis in vitro, as shown by tube formation and wound healing assays. Our data suggest a novel pharmacological strategy that enhances myocardial infarction-induced MSC mobilisation and improves cardiac function after myocardial infarction.
Collapse
Affiliation(s)
- Veneta B. Todorova
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| | - Nicoleta Baxan
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| | - Matthew Delahaye
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| | - Sian E. Harding
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| | - Sara M. Rankin
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| |
Collapse
|
27
|
Pham TLB, Nguyen DPH, Luu TTT, Nguyen LS, Binh NT, Nguyen QD, Tran PA. Encapsulation of Human Umbilical Cord Mesenchymal Stem Cells in LunaGel Photocrosslinkable Extracellular Matrix and Subcutaneous Transplantation in Mice. Biomedicines 2023; 11:biomedicines11041158. [PMID: 37189776 DOI: 10.3390/biomedicines11041158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Stem cells have significant potential in regenerative medicines. However, a major issue with implanting stem cells in the regeneration of new tissue is the methods to implant them and cell viability and functions before and after implantation. Here we developed a simple yet effective method that used photo-crosslinkable gelatin-based hydrogel (LunaGelTM) as a scaffold for the encapsulation, expansion, and eventually, transplantation of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) into mice subcutaneously. We demonstrated the proliferation and maintenance of the original expression of mesenchymal stem cell markers as well as the ability to differentiate into mesoderm-derived cells. The hydrogel was highly stable with no signs of degradation after 20 days in PBS. The hUC-MSCs remained viable after transplantation into mice's subcutaneous pockets and migrated to integrate with the surrounding tissues. We showed a collagen-rich layer surrounding the transplanted cell-laden scaffold indicating the effects of growth factors secreted by the hUC-MSCs. A connective tissue layer was found between the implanted cell-laden scaffold and the collagen layer, and immunohistochemical staining results suggested that this tissue was derived from the MSCs which migrated from within the scaffold. The results, thus, also suggested a protective effect the scaffold has on the encapsulated cells from the antibodies and cytotoxic cells of the host immune system.
Collapse
Affiliation(s)
- Truc Le-Buu Pham
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | | | - Thao Thi-Thu Luu
- Histology-Embryology-Pathology Department, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Luong Si Nguyen
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Nguyen Trong Binh
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Quan Dang Nguyen
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Phong Anh Tran
- Interface Science and Materials Engineering Group, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
| |
Collapse
|
28
|
Kilpatrick AM, Marques M, Gomez-Salazar M, Stefancova D, Sá da Bandeira D, Crisan M. Protocol to analyze and validate transcriptomic changes in PDGFRβ-KO mesenchymal stem cell osteogenic potential in the mouse embryo. STAR Protoc 2023; 4:102016. [PMID: 36640365 PMCID: PMC9846014 DOI: 10.1016/j.xpro.2022.102016] [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: 10/26/2022] [Revised: 12/02/2022] [Accepted: 12/22/2022] [Indexed: 01/14/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) can differentiate into osteoblasts under appropriate conditions. PDGFRβ signaling controls MSC osteogenic potential both transcriptomically and in culture. Here, we present a "computer to the bench" protocol to analyze changes in MSC osteogenic potential at transcriptomic and cellular level in the absence of PDGFRβ. We detail the preparation of cells from mouse embryos, the analysis of transcriptomic changes from single-cell RNA-sequencing data, the procedure for MSC derivation and culture, and an osteogenic assay for functional validation. For complete details on the use and execution of this protocol, please refer to Sá da Bandeira et al. (2022).1.
Collapse
Affiliation(s)
- Alastair Morris Kilpatrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Madalena Marques
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Mario Gomez-Salazar
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Dorota Stefancova
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Diana Sá da Bandeira
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK; Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Mihaela Crisan
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK; Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
29
|
Alves ED, Benevenuto LGD, Morais BP, Barros MA, Achcar JA, Montrezor LH. Ovarian Microenvironment Modulation by Adipose-Mesenchymal Stem Cells and Photobiomodulation Can Alter Osteoblasts Functions In Vitro. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2023. [DOI: 10.1007/s40883-023-00297-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
30
|
Bidkhori HR, Farshchian M, Kazemi Noughabi M, Hassanzadeh H, Rafatpanah H. Alteration of immunoregulatory genes expression in mesenchymal stromal cells upon priming with B18R as an interferon binding protein. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:241-247. [PMID: 36742146 PMCID: PMC9869884 DOI: 10.22038/ijbms.2022.67353.14771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/06/2022] [Indexed: 02/07/2023]
Abstract
Objectives The B18R protein encoded by the Vaccinia virus decoys Type 1 interferons and inhibits the activity of several type I IFN members. In vitro transcription protocols benefit from this molecule's involvement in enhancing cell viability by inhibiting interferon signal transduction. As a result of their immunomodulatory properties and potential to regenerate, mesenchymal stromal cells (MSCs) are increasingly considered an alternative treatment for a wide range of immune disorders. In this study, we investigated the modification of expression of several genes involved in immune-related pathways after preconditioning MSCs with two immune stimuli, including poly(I:C) and LPS. Materials and Methods ASCs were isolated and primed with B18R, and after exposure to poly(I:C) and LPS, the expression of the same sets of genes as in the previous experiment was evaluated. Following total RNA isolation from primed cells and cDNA preparation, real-time quantitative PCR was performed for several immunomodulatory and immune-related genes, including IDO1, TDO2, COX-2, TGF- β 1, TNF- α, IL-1 β , IL-6, TLR3, TLR4, and MCP-1. Results Pretreatment of MSCs with poly(I:C) and LPS significantly increased the expression of all mentioned genes, while upon the B18R challenge followed by poly(I:C) and LPS treatment, they were down-regulated. Finally, it was observed that the relative expression level of IFN -β has significantly decreased in MSCs+B18R+poly(I:C) and LPS in comparison with these groups without B18R. Conclusion The data indicated that the presence of B18R prevents the overexpression of several immune-related genes, which are overexpressed in the in vitro inflammatory environment.
Collapse
Affiliation(s)
- Hamid Reza Bidkhori
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Moein Farshchian
- Stem Cells and Regenerative Medicine Department, Academic Center for Education, Culture, and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | | | - Halimeh Hassanzadeh
- Stem Cells and Regenerative Medicine Department, Academic Center for Education, Culture, and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | - Houshang Rafatpanah
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran,Corresponding author: Houshang Rafatpanah. Immunology Research Center, Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Mashhad University of Medical Sciences, Azadi-Square, Medical Campus, Mashhad, Iran. Tel: +98-51-3 800 2376;
| |
Collapse
|
31
|
Quintero D, Perucca Orfei C, Kaplan LD, de Girolamo L, Best TM, Kouroupis D. The roles and therapeutic potentialof mesenchymal stem/stromal cells and their extracellular vesicles in tendinopathies. Front Bioeng Biotechnol 2023; 11:1040762. [PMID: 36741745 PMCID: PMC9892947 DOI: 10.3389/fbioe.2023.1040762] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/04/2023] [Indexed: 01/20/2023] Open
Abstract
Tendinopathies encompass a highly prevalent, multi-faceted spectrum of disorders, characterized by activity-related pain, compromised function, and propensity for an extended absence from sport and the workplace. The pathophysiology of tendinopathy continues to evolve. For decades, it has been related primarily to repetitive overload trauma but more recently, the onset of tendinopathy has been attributed to the tissue's failed attempt to heal after subclinical inflammatory and immune challenges (failed healing model). Conventional tendinopathy management produces only short-term symptomatic relief and often results in incomplete repair or healing leading to compromised tendon function. For this reason, there has been increased effort to develop therapeutics to overcome the tissue's failed healing response by targeting the cellular metaplasia and pro-inflammatory extra-cellular environment. On this basis, stem cell-based therapies have been proposed as an alternative therapeutic approach designed to modify the course of the various tendon pathologies. Mesenchymal stem/stromal cells (MSCs) are multipotent stem cells often referred to as "medicinal signaling cells" due to their immunomodulatory and anti-inflammatory properties that can produce a pro-regenerative microenvironment in pathological tendons. However, the adoption of MSCs into clinical practice has been limited by FDA regulations and perceived risk of adverse events upon infusion in vivo. The introduction of cell-free approaches, such as the extracellular vesicles of MSCs, has encouraged new perspectives for the treatment of tendinopathies, showing promising short-term results. In this article, we review the most recent advances in MSC-based and MSC-derived therapies for tendinopathies. Preclinical and clinical studies are included with comment on future directions of this rapidly developing therapeutic modality, including the importance of understanding tissue loading and its relationship to any treatment regimen.
Collapse
Affiliation(s)
- Daniel Quintero
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Carlotta Perucca Orfei
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Lee D. Kaplan
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Laura de Girolamo
- Laboratorio di Biotecnologie Applicate all’Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Thomas M. Best
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Dimitrios Kouroupis
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States,Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL, United States,*Correspondence: Dimitrios Kouroupis,
| |
Collapse
|
32
|
Ibrahim D, Abozied N, Abdel Maboud S, Alzamami A, Alturki NA, Jaremko M, Alanazi MK, Alhuthali HM, Seddek A. Therapeutic potential of bone marrow mesenchymal stem cells in cyclophosphamide-induced infertility. Front Pharmacol 2023; 14:1122175. [PMID: 37033609 PMCID: PMC10073512 DOI: 10.3389/fphar.2023.1122175] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/26/2023] [Indexed: 04/11/2023] Open
Abstract
Cancer is a deadly disease characterized by abnormal cell proliferation. Chemotherapy is one technique of cancer treatment. Cyclophosphamide (CYP) is the most powerful chemotherapy medication, yet it has serious adverse effects. It is an antimitotic medicine that regulates cell proliferation and primarily targets quickly dividing cells, and it has been related to varying levels of infertility in humans. In the current study, we assessed the biochemical, histological, and microscopic evaluations of testicular damage following cyclophosphamide administration. Further, we have explored the potential protective impact of mesenchymal stem cell (MSCs) transplantation. The biochemical results revealed that administration of cyclophosphamide increased serum concentrations of follicle-stimulating hormone (FSH) and luteinizing hormone (LH), while it decreased serum concentrations of free testosterone hormone (TH), testicular follicle-stimulating hormone, luteinizing hormone, and free testosterone hormone concentrations, testicular total antioxidant capacity (TAC), and testicular activity of superoxide dismutase (SOD) enzyme. The histology and sperm examinations revealed that cyclophosphamide induced destruction to the architectures of several tissues in the testes, which drastically reduced the Johnsen score as well as the spermatogenesis process. Surprisingly, transplantation of mesenchymal stem cell after cyclophosphamide administration altered the deterioration effect of cyclophosphamide injury on the testicular tissues, as demonstrated by biochemical and histological analysis. Our results indicated alleviation of serum and testicular sex hormones, as well as testicular oxidative stress markers (total antioxidant capacity and superoxide dismutase activity), and nearly restored the normal appearance of the testicular tissues, Johnsen score, and spermatogenesis process. In conclusion, our work emphasizes the protective pharmacological use of mesenchymal stem cell to mitigate the effects of cyclophosphamide on testicular tissues that impair the spermatogenesis process following chemotherapy. These findings indicate that transferring mesenchymal stem cell to chemotherapy patients could significantly improve spermatogenesis.
Collapse
Affiliation(s)
- Dalia Ibrahim
- The Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
- *Correspondence: Dalia Ibrahim,
| | - Nadia Abozied
- The Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Samar Abdel Maboud
- The Department of Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ahmad Alzamami
- Clinical Laboratory Science Department, College of Applied Medical Science, Shaqra University, AlQuwayiyah, Saudi Arabia
| | - Norah A. Alturki
- Clinical Laboratory Science Department, College of Applied Medical Science, King Saud University, Riyadh, Saudi Arabia
| | - Mariusz Jaremko
- Smart-Health Initiative and Red Sea Research Center, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Maram Khalil Alanazi
- Pharm.D, Scientific Office and Regulatory Affair Department, Dallah Pharma Company, Riyadh, Saudi Arabia
| | - Hayaa M. Alhuthali
- Department of Clinical laboratory sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Asmaa Seddek
- The Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
33
|
Alipour R, Hashemibeni B, Asgari V, Bahramian H. Time- and Concentration-Dependent Effects of the Stem Cells Derived from Human Exfoliated Deciduous Teeth on Osteosarcoma Cells. Adv Biomed Res 2023; 12:81. [PMID: 37200742 PMCID: PMC10186045 DOI: 10.4103/abr.abr_277_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/14/2021] [Accepted: 10/11/2021] [Indexed: 05/20/2023] Open
Abstract
Background Stem cells have been proposed to be one of the potent sources for treatment applications. Among diverse types of stem cells, stem cells derived from human exfoliated deciduous teeth (SHEDs) are known as the immature stem cell population, which are easily isolated, fast, and without ethical implications. SHEDs could induce pluripotent stem cells and show differentiation in chondrocytes, adipocytes, osteoblasts, neural cells, hepatocytes, myocytes, odontoblasts, and skin cells. Materials and Methods In the current study, we investigated the effects of SHED on osteosarcoma cells (Saos-II) following 3 and 5 days indirect coculture. Results Our results showed that indirect coculture of SHED with Saos-II cells could promote or inhibit Saos-II cells' growth in a concentration (the number of SHED vs. Saos-II cells) and time (days of indirect co-culture) dependent manner. Conclusion Our findings suggested that, indirectly, SHED co-culture with the Soas-II cells might functions as a tumor suppressor where a higher number of SHEDs are used in the culture in comparison with the one cultured in the absence of/or fewer SHED incubation.
Collapse
Affiliation(s)
- Razieh Alipour
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Batool Hashemibeni
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vajihe Asgari
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Bahramian
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Address for correspondence: Dr. Hamid Bahramian, Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran. E-mail:
| |
Collapse
|
34
|
Dabravolski SA, Markin AM, Andreeva ER, Eremin II, Orekhov AN, Melnichenko AA. Emerging role of pericytes in therapy of cardiovascular diseases. Biomed Pharmacother 2022; 156:113928. [DOI: 10.1016/j.biopha.2022.113928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/17/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
|
35
|
Stem cell sheet fabrication from human umbilical cord mesenchymal stem cell and Col-T scaffold. Stem Cell Res 2022; 65:102960. [PMID: 36399925 DOI: 10.1016/j.scr.2022.102960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Today, stem cell therapy has been shown to be a remarkable progress and an important application in the regeneration of defective tissues and organs. To deliver stem cells to the injured area, several methods have been proposed such as an intravenous infusion, direct damaged tissue injection, or stem cell sheet transplantation. In this study, we aimed to fabricate a stem cell sheet by culturing human umbilical cord mesenchymal stem cells (hUC-MSCs) on a Col-T scaffold to recover the structure and function of damaged tissues. The results showed that cells reach confluent on the scaffold surface 18 h after seeding. These stem cells were able to survive and proliferate on Col-T scaffold. The average tensile strength of the stem cell sheet was 2.65 MPa. The sheet reached the sterile standards when tested for total bacteria, Candida albicans, Pseudomonas aeruginosa, and Staphylococcus aureus according to Circular number 06/2011/TT-BYT of Vietnam Ministry of Health. In addition, the stem cell sheet was non-toxic when evaluated for exposure toxicity and fluid toxicity according to iSO-10993. Importantly, 5 days after culturing on the Col-T scaffold, the seeded hUC-MSCs were still possessed all properties of MSC such as spindle-shaped, adhesive, could differentiate into mesoderm-derived cells, showed to be CD90, CD105, CD73 positive and CD45, CD34, CD11b, CD19, HLA-DR negative. In summary, our study was successful in creating a stem cell sheet from hUC-MSCs and Col-T scaffold for subsequent in vivo transplantation in the future.
Collapse
|
36
|
Hamann A, Pannier AK. Innovative nonviral gene delivery strategies for engineering human mesenchymal stem cell phenotypes toward clinical applications. Curr Opin Biotechnol 2022; 78:102819. [PMID: 36274497 DOI: 10.1016/j.copbio.2022.102819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 12/14/2022]
Abstract
Although human mesenchymal stem cells (hMSCs) have been used in many clinical trials, variable outcomes have resulted in no FDA-approved hMSC treatment. However, research into developing hMSC therapies for many diseases continues. An approach to manipulate hMSCs for therapeutic applications is gene delivery. Nonviral gene delivery is safer and more flexible than viral vectors, but much less efficient, especially in hMSCs. It is not understood why hMSCs are more difficult to transfect than cell lines, but innate features of hMSCs may present unique barriers to transfection. Recently, strategies to improve hMSC transfection have been developed by innovating nanocarriers, nucleic acid cargos, and by 'priming' hMSCs chemically and physically for more efficient transfection. These strategies aim to engineer hMSCs with new phenotypes mediated by transgenic secreted factors, receptors, transcription factors, and genome editing systems for clinical applications requiring enhanced immunomodulation and/or tissue regeneration, or for functions such as tumor-killing and tissue engineering.
Collapse
Affiliation(s)
- Andrew Hamann
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Angela K Pannier
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
37
|
Pouikli A, Maleszewska M, Parekh S, Yang M, Nikopoulou C, Bonfiglio JJ, Mylonas C, Sandoval T, Schumacher A, Hinze Y, Matic I, Frezza C, Tessarz P. Hypoxia promotes osteogenesis by facilitating acetyl-CoA-mediated mitochondrial-nuclear communication. EMBO J 2022; 41:e111239. [PMID: 36278281 PMCID: PMC9713713 DOI: 10.15252/embj.2022111239] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 09/17/2022] [Accepted: 09/23/2022] [Indexed: 01/15/2023] Open
Abstract
Bone-derived mesenchymal stem cells (MSCs) reside in a hypoxic niche that maintains their differentiation potential. While hypoxia (low oxygen concentration) was reported to critically support stem cell function and osteogenesis, the molecular events triggering changes in stem cell fate decisions in response to normoxia (high oxygen concentration) remain elusive. Here, we study the impact of normoxia on mitochondrial-nuclear communication during stem cell differentiation. We show that normoxia-cultured murine MSCs undergo profound transcriptional alterations which cause irreversible osteogenesis defects. Mechanistically, high oxygen promotes chromatin compaction and histone hypo-acetylation, particularly on promoters and enhancers of osteogenic genes. Although normoxia induces metabolic rewiring resulting in elevated acetyl-CoA levels, histone hypo-acetylation occurs due to the trapping of acetyl-CoA inside mitochondria owing to decreased citrate carrier (CiC) activity. Restoring the cytosolic acetyl-CoA pool remodels the chromatin landscape and rescues the osteogenic defects. Collectively, our results demonstrate that the metabolism-chromatin-osteogenesis axis is perturbed upon exposure to high oxygen levels and identifies CiC as a novel, oxygen-sensitive regulator of the MSC function.
Collapse
Affiliation(s)
- Andromachi Pouikli
- Max Planck Research Group “Chromatin and Ageing”Max Planck Institute for Biology of AgeingCologneGermany
- Cologne Excellence Cluster on Stress Responses in Ageing‐Associated Diseases (CECAD)CologneGermany
| | - Monika Maleszewska
- Max Planck Research Group “Chromatin and Ageing”Max Planck Institute for Biology of AgeingCologneGermany
- Present address:
CareDx, Inc.San FranciscoCAUSA
| | - Swati Parekh
- Max Planck Research Group “Chromatin and Ageing”Max Planck Institute for Biology of AgeingCologneGermany
| | - Ming Yang
- Cologne Excellence Cluster on Stress Responses in Ageing‐Associated Diseases (CECAD)CologneGermany
| | - Chrysa Nikopoulou
- Max Planck Research Group “Chromatin and Ageing”Max Planck Institute for Biology of AgeingCologneGermany
| | - Juan Jose Bonfiglio
- Research Group “Proteomics and ADP‐Ribosylation Signaling”Max Planck Institute for Biology of AgeingCologneGermany
- Present address:
Roche Pharma Research and Early DevelopmentMunichGermany
| | - Constantine Mylonas
- Max Planck Research Group “Chromatin and Ageing”Max Planck Institute for Biology of AgeingCologneGermany
- Present address:
Novartis Institutes for BioMedical ResearchCambridgeMAUSA
| | - Tonantzi Sandoval
- Max Planck Research Group “Chromatin and Ageing”Max Planck Institute for Biology of AgeingCologneGermany
| | - Anna‐Lena Schumacher
- FACS & Imaging Core FacilityMax Planck Institute for Biology of AgeingCologneGermany
| | - Yvonne Hinze
- Metabolomics Core Facility, Max Planck Institute for Biology of AgeingCologneGermany
| | - Ivan Matic
- Cologne Excellence Cluster on Stress Responses in Ageing‐Associated Diseases (CECAD)CologneGermany
- Research Group “Proteomics and ADP‐Ribosylation Signaling”Max Planck Institute for Biology of AgeingCologneGermany
| | - Christian Frezza
- Cologne Excellence Cluster on Stress Responses in Ageing‐Associated Diseases (CECAD)CologneGermany
| | - Peter Tessarz
- Max Planck Research Group “Chromatin and Ageing”Max Planck Institute for Biology of AgeingCologneGermany
- Cologne Excellence Cluster on Stress Responses in Ageing‐Associated Diseases (CECAD)CologneGermany
| |
Collapse
|
38
|
Akasaka Y. The Role of Mesenchymal Stromal Cells in Tissue Repair and Fibrosis. Adv Wound Care (New Rochelle) 2022; 11:561-574. [PMID: 34841889 DOI: 10.1089/wound.2021.0037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Significance: The present review covers an overview of the current understanding of biology of mesenchymal stromal cells (MSCs) and suggests an important role of their differential potential for clinical approaches associated with tissue repair and fibrosis. Recent Advances: Genetic lineage tracing technology has enabled the delineation of cellular hierarchies and examination of MSC cellular origins and myofibroblast sources. This technique has led to the characterization of perivascular MSC populations and suggests that pericytes might provide a local source of tissue-specific MSCs, which can differentiate into tissue-specific cells for tissue repair and fibrosis. Autologous adipose tissue MSCs led to the advance in tissue engineering for regeneration of damaged tissues. Critical Issues: Recent investigation has revealed that perivascular MSCs might be the origin of myofibroblasts during fibrosis development, and perivascular MSCs might be the major source of myofibroblasts in fibrogenic disease. Adipose tissue MSCs combined with cytokines and biomaterials are available in the treatment of soft tissue defect and skin wound healing. Future Directions: Further investigation of the roles of perivascular MSCs may enable new approaches in the treatment of fibrogenic disease; moreover, perivascular MSCs might have potential as an antifibrotic target for fibrogenic disease. Autologous adipose tissue MSCs combined with cytokines and biomaterials will be an alternative method for the treatment of soft tissue defect and skin wound healing.
Collapse
Affiliation(s)
- Yoshikiyo Akasaka
- Division of Research Promotion and Development, Advanced Research Center, Toho University Graduate School of Medicine, Ota-ku, Japan.,Department of Pathology, Toho University School of Medicine, Ota-ku, Japan
| |
Collapse
|
39
|
Osteogenic Differentiation of Periodontal Ligament Stem Cells Seeded on Equine-Derived Xenograft in Osteogenic Growth Media. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58111518. [PMID: 36363474 PMCID: PMC9693579 DOI: 10.3390/medicina58111518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/17/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Background and Objectives: The duration of bone turnover is critical, and different time points help in identifying the optimal endpoint of treatment duration. However, investigating the combination of xenograft and stem cells to allow tissue regeneration within an ideal time duration remains an under-investigated topic. The current study aimed to assess the impact of equine-derived xenograft bone blocks in assisting the human periodontal ligament stem cells (PDLSCs) to demonstrate osteogenic differentiation (collagen type 1 expression and calcium deposition) within an osteogenic growth media. Materials and Methods: Human PDLSCs were acquired commercially and seeded onto xenograft bone blocks. After the 14th and 21st day of culture, enzyme-linked immunoassay (ELISA) was utilized for the detection and quantification of levels of collagen type I, while the mineralization assessment (deposition of calcium) was conducted by staining the PDLSCs with Alizarin Red S (ARS). The statistical comparison between the means and standard deviations of study groups were evaluated using analysis of variance (ANOVA). Results: ELISA assessment revealed an upsurge in the expression of collagen type I for PDLSCs cultured with xenograft after 14 and 21 days compared to the controls (intergroup comparisons significant at p < 0.05). Similar findings were obtained for mineralization assessment and on ARS staining. PDLSCs cultured with xenograft bone blocks presented an increased deposition of calcium compared to their control counterparts (intergroup comparisons significant at p < 0.05). Conclusions: PDLSCs embedded in xenograft bone blocks inside an osteogenic growth medium demonstrated greater osteogenic differentiation potential after 14 and 21 days. This superior osteogenic differentiation capability was evident by increased collagen type I expression and more significant calcium deposition at the 14th and 21st days after culture.
Collapse
|
40
|
Yoshimura TM, Cabral FV, Sellera FP, Pozzo L, Ribeiro MS. Could Light-Based Technologies Improve Stem Cell Therapy for Skin Wounds? A Systematic Review and Meta-Analysis of Preclinical Studies. Photochem Photobiol 2022; 99:519-528. [PMID: 36004458 DOI: 10.1111/php.13702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/22/2022] [Indexed: 12/09/2022]
Abstract
Several diseases or conditions cause dermatological disorders that hinder the process of skin repair. The search for novel technologies has inspired the combination of stem cell (SC) and light-based therapies to ameliorate skin wound repair. Herein, we systematically revised the impact of photobiomodulation therapy (PBM) combined with SCs in animal models of skin wounds and quantitatively evaluated this effect through a meta-analysis. For inclusion, SCs should be irradiated in vitro or in vivo, before or after being implanted in animals, respectively. The search resulted in nine eligible articles, which were assessed for risk of bias. For the meta-analysis, studies were included only when PBM was applied in vivo, five regarding wound closure, and three to wound strength. Overall, a positive influence of SC+PBM on wound closure (MD: 9.69; 95%CI: 5.78 to 13.61, p<0.00001) and strength (SMD: 1.7, 95%CI: 0.68 to 2.72, p=0.001) was detected, although studies have shown moderate to high heterogeneity and a lack of information regarding some bias domains. Altogether, PBM seems to be an enabling technology able to be applied post-implantation of SCs for cutaneous regeneration. Our findings may guide future laboratory and clinical studies in hopes of offering wound care patients a better quality of life.
Collapse
Affiliation(s)
- Tania M Yoshimura
- Center for Lasers and Applications, Nuclear and Energy Research Institute (IPEN-CNEN), São Paulo, SP, Brazil
| | - Fernanda V Cabral
- Center for Lasers and Applications, Nuclear and Energy Research Institute (IPEN-CNEN), São Paulo, SP, Brazil
| | - Fábio P Sellera
- Department of Internal Medicine, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil.,School of Veterinary Medicine, Metropolitan University of Santos, Santos, SP, Brazil
| | - Lorena Pozzo
- Health Technology Assessment Nucleus, Nuclear and Energy Research Institute (IPEN-CNEN), São Paulo, SP, Brazil
| | - Martha S Ribeiro
- Center for Lasers and Applications, Nuclear and Energy Research Institute (IPEN-CNEN), São Paulo, SP, Brazil.,Health Technology Assessment Nucleus, Nuclear and Energy Research Institute (IPEN-CNEN), São Paulo, SP, Brazil
| |
Collapse
|
41
|
Malinauskas M, Jankauskaite L, Aukstikalne L, Dabasinskaite L, Rimkunas A, Mickevicius T, Pockevicius A, Krugly E, Martuzevicius D, Ciuzas D, Baniukaitiene O, Usas A. Cartilage regeneration using improved surface electrospun bilayer polycaprolactone scaffolds loaded with transforming growth factor-beta 3 and rabbit muscle-derived stem cells. Front Bioeng Biotechnol 2022; 10:971294. [PMID: 36082160 PMCID: PMC9445302 DOI: 10.3389/fbioe.2022.971294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/19/2022] [Indexed: 11/15/2022] Open
Abstract
Polycaprolactone (PCL) has recently received significant attention due to its mechanical strength, low immunogenicity, elasticity, and biodegradability. Therefore, it is perfectly suitable for cartilage tissue engineering. PCL is relatively hydrophobic in nature, so its hydrophilicity needs to be enhanced before its use in scaffolding. In our study, first, we aimed to improve the hydrophilicity properties after the network of the bilayer scaffold was formed by electrospinning. Electrospun bilayer PCL scaffolds were treated with ozone and further loaded with transforming growth factor-beta 3 (TGFβ3). In vitro studies were performed to determine the rabbit muscle-derived stem cells’ (rMDSCs) potential to differentiate into chondrocytes after the cells were seeded onto the scaffolds. Statistically significant results indicated that ozonated (O) scaffolds create a better environment for rMDSCs because collagen-II (Coll2) concentrations at day 21 were higher than non-ozonated (NO) scaffolds. In in vivo studies, we aimed to determine the cartilage regeneration outcomes by macroscopical and microscopical/histological evaluations at 3- and 6-month time-points. The Oswestry Arthroscopy Score (OAS) was the highest at both mentioned time-points using the scaffold loaded with TGFβ3 and rMDSCs. Evaluation of cartilage electromechanical quantitative parameters (QPs) showed significantly better results in cell-treated scaffolds at both 3 and 6 months. Safranin O staining indicated similar results as in macroscopical evaluations—cell-treated scaffolds revealed greater staining with safranin, although an empty defect also showed better results than non-cell-treated scaffolds. The scaffold with chondrocytes represented the best score when the scaffolds were evaluated with the Mankin histological grading scale. However, as in previous in vivo evaluations, cell-treated scaffolds showed better results than non-cell-treated scaffolds. In conclusion, we have investigated that an ozone-treated scaffold containing TGFβ3 with rMDSC is a proper combination and could be a promising scaffold for cartilage regeneration.
Collapse
Affiliation(s)
- Mantas Malinauskas
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- *Correspondence: Mantas Malinauskas,
| | - Lina Jankauskaite
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Lauryna Aukstikalne
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | - Augustinas Rimkunas
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Tomas Mickevicius
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Alius Pockevicius
- Department of Veterinary Pathobiology, Veterinary Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Edvinas Krugly
- Faculty of Chemical Technology, Kaunas University of Technology, Kaunas, Lithuania
| | | | - Darius Ciuzas
- Faculty of Chemical Technology, Kaunas University of Technology, Kaunas, Lithuania
| | - Odeta Baniukaitiene
- Faculty of Chemical Technology, Kaunas University of Technology, Kaunas, Lithuania
| | - Arvydas Usas
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
42
|
Krishna M, Shetty A, Manjappa AB, Shetty V, Hegde MN, Kumar BM. Comparative characterization and analysis of telomere length in stem cells derived from deciduous and permanent teeth. Dent Res J (Isfahan) 2022; 19:64. [PMID: 36159052 PMCID: PMC9490240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 01/24/2022] [Accepted: 03/01/2022] [Indexed: 11/18/2022] Open
Abstract
Background Understanding the influence of age on growth kinetics and telomere length in dental stem cells is essential for the successful development of cell therapies. Hence, the present study compared the basic cellular and phenotypical characteristics of stem cells from human exfoliated deciduous teeth (SHEDs) and dental pulp stem cells (DPSCs) of permanent teeth and their telomere lengths using quantitative real-time polymerase chain reaction. Materials and Methods The study is an in vitro original research article. Primary cultures of SHED and DPSCs (n = 6 each) were successfully established in vitro, and the parameters analyzed were the morphology, viability, proliferation rate, population doubling time (PDT), phenotypic markers expression, and the relative telomere lengths. Data were analyzed by analysis of variance and P < 0.05 was considered statistically significant. Results SHED and DPSCs exhibited a small spindle-shaped fibroblast-like morphology with >90% viability. The proliferation assay showed that the cells had a typical growth pattern. The PDT values of SHED and DPSCs were 29.03 ± 9.71 h and 32.05 ± 9.76 h, respectively. Both cells were positive for surface markers CD29, CD44, and CD90. However, they were negative for CD45 and human leukocyte antigen DR. Although the differences in relative telomere lengths between the individual cell lines of SHED and DPSCs were observed, no significant (P > 0.05) variations were found for the mean T/S ratios of both the cells. Conclusion SHED and DPSCs displayed similar morphology, proliferation rates, and phenotypic features. The relative telomere lengths were slightly shorter in DPSCs than SHED, but the values were not significantly different. Thus, SHED and DPSCs can be considered as recognized sources for regenerative applications in dentistry.
Collapse
Affiliation(s)
- Murali Krishna
- Department of Conservative Dentistry and Endodontics, AB Shetty Memorial Institute of Dental Sciences, Nitte (Deemed to be University), Mangaluru, Karnataka, India
| | - Aditya Shetty
- Department of Conservative Dentistry and Endodontics, AB Shetty Memorial Institute of Dental Sciences, Nitte (Deemed to be University), Mangaluru, Karnataka, India,Address for correspondence: Dr. Aditya Shetty, Department of Conservative Dentistry and Endodontics, AB Shetty Memorial Institute of Dental Sciences, Nitte (Deemed to be University), Mangaluru, Karnataka, India. E-mail:
| | - Akshay Bairapura Manjappa
- Nitte University Centre for Stem Cell Research and Regenerative Medicine, KS Hegde Medical Academy, Nitte (Deemed to be University), Mangaluru, Karnataka, India
| | - Veena Shetty
- Nitte University Centre for Stem Cell Research and Regenerative Medicine, KS Hegde Medical Academy, Nitte (Deemed to be University), Mangaluru, Karnataka, India
| | - Mithra Nidarsh Hegde
- Department of Conservative Dentistry and Endodontics, AB Shetty Memorial Institute of Dental Sciences, Nitte (Deemed to be University), Mangaluru, Karnataka, India
| | - Basavarajappa Mohana Kumar
- Nitte University Centre for Stem Cell Research and Regenerative Medicine, KS Hegde Medical Academy, Nitte (Deemed to be University), Mangaluru, Karnataka, India
| |
Collapse
|
43
|
Tan MI, Alfarafisa NM, Septiani P, Barlian A, Firmansyah M, Faizal A, Melani L, Nugrahapraja H. Potential Cell-Based and Cell-Free Therapy for Patients with COVID-19. Cells 2022; 11:2319. [PMID: 35954162 PMCID: PMC9367488 DOI: 10.3390/cells11152319] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 02/01/2023] Open
Abstract
Since it was first reported, the novel coronavirus disease 2019 (COVID-19) remains an unresolved puzzle for biomedical researchers in different fields. Various treatments, drugs, and interventions were explored as treatments for COVID. Nevertheless, there are no standard and effective therapeutic measures. Meanwhile, mesenchymal stem cell (MSC) therapy offers a new approach with minimal side effects. MSCs and MSC-based products possess several biological properties that potentially alleviate COVID-19 symptoms. Generally, there are three classifications of stem cell therapy: cell-based therapy, tissue engineering, and cell-free therapy. This review discusses the MSC-based and cell-free therapies for patients with COVID-19, their potential mechanisms of action, and clinical trials related to these therapies. Cell-based therapies involve the direct use and injection of MSCs into the target tissue or organ. On the other hand, cell-free therapy uses secreted products from cells as the primary material. Cell-free therapy materials can comprise cell secretomes and extracellular vesicles. Each therapeutic approach possesses different benefits and various risks. A better understanding of MSC-based and cell-free therapies is essential for supporting the development of safe and effective COVID-19 therapy.
Collapse
Affiliation(s)
- Marselina Irasonia Tan
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung 40132, Indonesia; (P.S.); (A.B.); (M.F.); (A.F.); (L.M.); (H.N.)
| | - Nayla Majeda Alfarafisa
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang 45363, Indonesia;
| | - Popi Septiani
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung 40132, Indonesia; (P.S.); (A.B.); (M.F.); (A.F.); (L.M.); (H.N.)
| | - Anggraini Barlian
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung 40132, Indonesia; (P.S.); (A.B.); (M.F.); (A.F.); (L.M.); (H.N.)
| | - Mochamad Firmansyah
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung 40132, Indonesia; (P.S.); (A.B.); (M.F.); (A.F.); (L.M.); (H.N.)
| | - Ahmad Faizal
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung 40132, Indonesia; (P.S.); (A.B.); (M.F.); (A.F.); (L.M.); (H.N.)
| | - Lili Melani
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung 40132, Indonesia; (P.S.); (A.B.); (M.F.); (A.F.); (L.M.); (H.N.)
| | - Husna Nugrahapraja
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung 40132, Indonesia; (P.S.); (A.B.); (M.F.); (A.F.); (L.M.); (H.N.)
| |
Collapse
|
44
|
Lebeau G, Ah-Pine F, Daniel M, Bedoui Y, Vagner D, Frumence E, Gasque P. Perivascular Mesenchymal Stem/Stromal Cells, an Immune Privileged Niche for Viruses? Int J Mol Sci 2022; 23:ijms23148038. [PMID: 35887383 PMCID: PMC9317325 DOI: 10.3390/ijms23148038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/16/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) play a critical role in response to stress such as infection. They initiate the removal of cell debris, exert major immunoregulatory activities, control pathogens, and lead to a remodeling/scarring phase. Thus, host-derived ‘danger’ factors released from damaged/infected cells (called alarmins, e.g., HMGB1, ATP, DNA) as well as pathogen-associated molecular patterns (LPS, single strand RNA) can activate MSCs located in the parenchyma and around vessels to upregulate the expression of growth factors and chemoattractant molecules that influence immune cell recruitment and stem cell mobilization. MSC, in an ultimate contribution to tissue repair, may also directly trans- or de-differentiate into specific cellular phenotypes such as osteoblasts, chondrocytes, lipofibroblasts, myofibroblasts, Schwann cells, and they may somehow recapitulate their neural crest embryonic origin. Failure to terminate such repair processes induces pathological scarring, termed fibrosis, or vascular calcification. Interestingly, many viruses and particularly those associated to chronic infection and inflammation may hijack and polarize MSC’s immune regulatory activities. Several reports argue that MSC may constitute immune privileged sanctuaries for viruses and contributing to long-lasting effects posing infectious challenges, such as viruses rebounding in immunocompromised patients or following regenerative medicine therapies using MSC. We will herein review the capacity of several viruses not only to infect but also to polarize directly or indirectly the functions of MSC (immunoregulation, differentiation potential, and tissue repair) in clinical settings.
Collapse
Affiliation(s)
- Grégorie Lebeau
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
| | - Franck Ah-Pine
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Service Anatomo-Pathologie, CHU de la Réunion, 97400 Saint-Denis, France
| | - Matthieu Daniel
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
| | - Yosra Bedoui
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
| | - Damien Vagner
- Service de Médecine Interne, CHU de la Réunion, 97400 Saint-Denis, France;
| | - Etienne Frumence
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
| | - Philippe Gasque
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, 97400 Saint-Denis, France; (G.L.); (F.A.-P.); (M.D.); (Y.B.); (E.F.)
- Laboratoire d’Immunologie Clinique et Expérimentale de la ZOI (LICE-OI), Pôle de Biologie, CHU de La Réunion, 97400 Saint-Denis, France
- Correspondence:
| |
Collapse
|
45
|
Dave C, Mei SHJ, McRae A, Hum C, Sullivan KJ, Champagne J, Ramsay T, McIntyre L. Comparison of freshly cultured versus cryopreserved mesenchymal stem cells in animal models of inflammation: A pre-clinical systematic review. eLife 2022; 11:75053. [PMID: 35838024 PMCID: PMC9286731 DOI: 10.7554/elife.75053] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 06/05/2022] [Indexed: 12/09/2022] Open
Abstract
Background: Mesenchymal stem cells (MSCs) are multipotent cells that demonstrate therapeutic potential for the treatment of acute and chronic inflammatory-mediated conditions. Although controversial, some studies suggest that MSCs may lose their functionality with cryopreservation which could render them non-efficacious. Hence, we conducted a systematic review of comparative pre-clinical models of inflammation to determine if there are differences in in vivo measures of pre-clinical efficacy (primary outcomes) and in vitro potency (secondary outcomes) between freshly cultured and cryopreserved MSCs. Methods: A systematic search on OvidMEDLINE, EMBASE, BIOSIS, and Web of Science (until January 13, 2022) was conducted. The primary outcome included measures of in vivo pre-clinical efficacy; secondary outcomes included measures of in vitro MSC potency. Risk of bias was assessed by the SYRCLE ‘Risk of Bias’ assessment tool for pre-clinical studies. Results: Eighteen studies were included. A total of 257 in vivo pre-clinical efficacy experiments represented 101 distinct outcome measures. Of these outcomes, 2.3% (6/257) were significantly different at the 0.05 level or less; 2 favoured freshly cultured and 4 favoured cryopreserved MSCs. A total of 68 in vitro experiments represented 32 different potency measures; 13% (9/68) of the experiments were significantly different at the 0.05 level or less, with seven experiments favouring freshly cultured MSC and two favouring cryopreserved MSCs. Conclusions: The majority of preclinical primary in vivo efficacy and secondary in vitro potency outcomes were not significantly different (p<0.05) between freshly cultured and cryopreserved MSCs. Our systematic summary of the current evidence base may provide MSC basic and clinical research scientists additional rationale for considering a cryopreserved MSC product in their pre-clinical studies and clinical trials as well as help identify research gaps and guide future related research. Funding: Ontario Institute for Regenerative Medicine
Collapse
Affiliation(s)
- Chintan Dave
- Division of Critical Care Medicine, Department of Medicine, Western University, London, Canada
| | - Shirley H J Mei
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Andrea McRae
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Christine Hum
- Knowledge Synthesis Group, Ottawa Hospital Research Institute, Ottawa, Canada.,University of Ottawa, Ottawa, Canada
| | - Katrina J Sullivan
- Knowledge Synthesis Group, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Josee Champagne
- Knowledge Synthesis Group, Ottawa Hospital Research Institute, Ottawa, Canada.,Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Tim Ramsay
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Lauralyn McIntyre
- Knowledge Synthesis Group, Ottawa Hospital Research Institute, Ottawa, Canada.,Division of Critical Care, Department of Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
46
|
Tamouza R, Volt F, Richard JR, Wu CL, Bouassida J, Boukouaci W, Lansiaux P, Cappelli B, Scigliuolo GM, Rafii H, Kenzey C, Mezouad E, Naamoune S, Chami L, Lejuste F, Farge D, Gluckman E. Possible Effect of the use of Mesenchymal Stromal Cells in the Treatment of Autism Spectrum Disorders: A Review. Front Cell Dev Biol 2022; 10:809686. [PMID: 35865626 PMCID: PMC9294632 DOI: 10.3389/fcell.2022.809686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
Autism spectrum disorder (ASD) represents a set of heterogeneous neurodevelopmental conditions defined by impaired social interactions and repetitive behaviors. The number of reported cases has increased over the past decades, and ASD is now a major public health burden. So far, only treatments to alleviate symptoms are available, with still unmet need for an effective disease treatment to reduce ASD core symptoms. Genetic predisposition alone can only explain a small fraction of the ASD cases. It has been reported that environmental factors interacting with specific inter-individual genetic background may induce immune dysfunctions and contribute to the incidence of ASD. Such dysfunctions can be observed at the central level, with increased microglial cells and activation in ASD brains or in the peripheral blood, as reflected by high circulating levels of pro-inflammatory cytokines, abnormal activation of T-cell subsets, presence of auto-antibodies and of dysregulated microbiota profiles. Altogether, the dysfunction of immune processes may result from immunogenetically-determined inefficient immune responses against a given challenge followed by chronic inflammation and autoimmunity. In this context, immunomodulatory therapies might offer a valid therapeutic option. Mesenchymal stromal cells (MSC) immunoregulatory and immunosuppressive properties constitute a strong rationale for their use to improve ASD clinical symptoms. In vitro studies and pre-clinical models have shown that MSC can induce synapse formation and enhance synaptic function with consequent improvement of ASD-like symptoms in mice. In addition, two preliminary human trials based on the infusion of cord blood-derived MSC showed the safety and tolerability of the procedure in children with ASD and reported promising clinical improvement of core symptoms. We review herein the immune dysfunctions associated with ASD provided, the rationale for using MSC to treat patients with ASD and summarize the current available studies addressing this subject.
Collapse
Affiliation(s)
- Ryad Tamouza
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
- *Correspondence: Ryad Tamouza,
| | - Fernanda Volt
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Jean-Romain Richard
- Translational Neuropsychiatry, INSERM, IMRB, Univ Paris Est Créteil, Créteil, France
| | - Ching-Lien Wu
- Translational Neuropsychiatry, INSERM, IMRB, Univ Paris Est Créteil, Créteil, France
| | - Jihène Bouassida
- Translational Neuropsychiatry, INSERM, IMRB, Univ Paris Est Créteil, Créteil, France
| | - Wahid Boukouaci
- Translational Neuropsychiatry, INSERM, IMRB, Univ Paris Est Créteil, Créteil, France
| | - Pauline Lansiaux
- Unité de Médecine Interne (UF 04), CRMR MATHEC, Maladies Auto-immunes et Thérapie Cellulaire, Centre de Référence des Maladies Auto-immunes Systémiques Rares D’Ile-de-France MATHEC, AP-HP, Hôpital St-Louis, Paris, France
| | - Barbara Cappelli
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- Monacord, Centre Scientifique de Monaco, Monaco, Monaco
| | - Graziana Maria Scigliuolo
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- Monacord, Centre Scientifique de Monaco, Monaco, Monaco
| | - Hanadi Rafii
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Chantal Kenzey
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Esma Mezouad
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
| | - Soumia Naamoune
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
| | - Leila Chami
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
| | - Florian Lejuste
- Translational Neuropsychiatry, INSERM, IMRB, DMU, AP-HP, Univ Paris Est Créteil, Créteil, France
| | - Dominique Farge
- Unité de Médecine Interne (UF 04), CRMR MATHEC, Maladies Auto-immunes et Thérapie Cellulaire, Centre de Référence des Maladies Auto-immunes Systémiques Rares D’Ile-de-France MATHEC, AP-HP, Hôpital St-Louis, Paris, France
| | - Eliane Gluckman
- Institut de Recherche Saint Louis (IRSL), Eurocord, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- Monacord, Centre Scientifique de Monaco, Monaco, Monaco
| |
Collapse
|
47
|
Moonshi SS, Adelnia H, Wu Y, Ta HT. Placenta‐Derived Mesenchymal Stem Cells for Treatment of Diseases: A Clinically Relevant Source. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Shehzahdi S. Moonshi
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
| | - Hossein Adelnia
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St Lucia Queensland 4072 Australia
| | - Yuao Wu
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
| | - Hang T. Ta
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
- Bioscience Discipline School of Environment and Science Griffith University Nathan Queensland 4111 Australia
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St Lucia Queensland 4072 Australia
| |
Collapse
|
48
|
Keshavarz Shahbaz S, Mansourabadi AH, Jafari D. Genetically engineered mesenchymal stromal cells as a new trend for treatment of severe acute graft-versus-host disease. Clin Exp Immunol 2022; 208:12-24. [PMID: 35274673 PMCID: PMC9113247 DOI: 10.1093/cei/uxac016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/25/2021] [Accepted: 02/07/2022] [Indexed: 01/12/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a population of non-hematopoietic and self-renewing cells characterized by the potential to differentiate into different cell subtypes. MSCs have interesting features which have attracted a lot of attention in various clinical investigations. Some basic features of MSCs are including the weak immunogenicity (absence of MHC-II and costimulatory ligands accompanied by the low expression of MHC-I) and the potential of plasticity and multi-organ homing via expressing related surface molecules. MSCs by immunomodulatory effects could also ameliorate several immune-pathological conditions like graft-versus-host diseases (GVHD). The efficacy and potency of MSCs are the main objections of MSCs therapeutic applications. It suggested that improving the MSC immunosuppressive characteristic via genetic engineering to produce therapeutic molecules consider as one of the best options for this purpose. In this review, we explain the functions, immunologic properties, and clinical applications of MSCs to discuss the beneficial application of genetically modified MSCs in GVHD.
Collapse
Affiliation(s)
- Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-communicable Disease, Qazvin University of Medical Science, Qazvin, Iran
| | - Amir Hossein Mansourabadi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Immunogenetics Research Network (IgReN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Davood Jafari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Immunogenetics Research Network (IgReN), Universal Scientific Education and Research Network (USERN), Zanjan, Iran
| |
Collapse
|
49
|
Brooks B, Ebedes D, Usmani A, Gonzales-Portillo JV, Gonzales-Portillo D, Borlongan CV. Mesenchymal Stromal Cells in Ischemic Brain Injury. Cells 2022; 11:cells11061013. [PMID: 35326464 PMCID: PMC8947674 DOI: 10.3390/cells11061013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 02/07/2023] Open
Abstract
Ischemic brain injury represents a major cause of death worldwide with limited treatment options with a narrow therapeutic window. Accordingly, novel treatments that extend the treatment from the early neuroprotective stage to the late regenerative phase may accommodate a much larger number of stroke patients. To this end, stem cell-based regenerative therapies may address this unmet clinical need. Several stem cell therapies have been tested as potentially exhibiting the capacity to regenerate the stroke brain. Based on the long track record and safety profile of transplantable stem cells for hematologic diseases, bone marrow-derived mesenchymal stromal cells or mesenchymal stromal cells have been widely tested in stroke animal models and have reached clinical trials. However, despite the translational promise of MSCs, probing cell function remains to be fully elucidated. Recognizing the multi-pronged cell death and survival processes that accompany stroke, here we review the literature on MSC definition, characterization, and mechanism of action in an effort to gain a better understanding towards optimizing its applications and functional outcomes in stroke.
Collapse
Affiliation(s)
- Beverly Brooks
- Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA; (B.B.); (D.E.); (A.U.)
| | - Dominique Ebedes
- Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA; (B.B.); (D.E.); (A.U.)
| | - Ahsan Usmani
- Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA; (B.B.); (D.E.); (A.U.)
| | | | | | - Cesario V. Borlongan
- Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA; (B.B.); (D.E.); (A.U.)
- Correspondence: ; Tel.: +1-8139743988
| |
Collapse
|
50
|
Jiang Z, Lin FY, Jiang K, Nguyen H, Chang CY, Lin CC. Dissolvable microgel-templated macroporous hydrogels for controlled cell assembly. BIOMATERIALS ADVANCES 2022; 134:112712. [PMID: 35581097 PMCID: PMC9358784 DOI: 10.1016/j.msec.2022.112712] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/05/2021] [Accepted: 02/08/2022] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cells (MSCs)-based therapies have been widely used to promote tissue regeneration and to modulate immune/inflammatory response. The therapeutic potential of MSCs can be further improved by forming multi-cellular spheroids. Meanwhile, hydrogels with macroporous structures are advantageous for improving mass transport properties for the cell-laden matrices. Herein, we report the fabrication of MSC-laden macroporous hydrogel scaffolds through incorporating rapidly dissolvable spherical cell-laden microgels. Dissolvable microgels were fabricated by tandem droplet-microfluidics and thiol-norbornene photopolymerization using a novel fast-degrading macromer poly(ethylene glycol)-norbornene-dopamine (PEGNB-Dopa). The cell-laden PEGNB-Dopa microgels were subsequently encapsulated within another bulk hydrogel matrix, whose porous structure was generated efficiently by the rapid degradation of the PEGNB-Dopa microgels. The cytocompatibility of this in situ pore-forming approach was demonstrated with multiple cell types. Furthermore, adjusting the stiffness and cell adhesiveness of the bulk hydrogels afforded the formation of solid cell spheroids or hollow spheres. The assembly of solid or hollow MSC spheroids led to differential activation of AKT pathway. Finally, MSCs solid spheroids formed in situ within the macroporous hydrogels exhibited robust secretion of HGF, VEGF-A, IL-6, IL-8, and TIMP-2. In summary, this platform provides an innovative method for forming cell-laden macroporous hydrogels for a variety of future biomedical applications.
Collapse
Affiliation(s)
- Zhongliang Jiang
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202 USA
| | - Fang-Yi Lin
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202 USA
| | - Kun Jiang
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202 USA
| | - Han Nguyen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
| | - Chun-Yi Chang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
| | - Chien-Chi Lin
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA.
| |
Collapse
|