1
|
Giebel B, Lim SK. Overcoming challenges in MSC-sEV therapeutics: insights and advances after a decade of research. Cytotherapy 2025:S1465-3249(25)00591-2. [PMID: 40243980 DOI: 10.1016/j.jcyt.2025.03.505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/14/2025] [Accepted: 03/14/2025] [Indexed: 04/18/2025]
Abstract
Over the past decade, mesenchymal stromal cell-derived small extracellular vesicles (MSC-sEVs) have emerged as promising therapeutics, shifting the focus from MSC engraftment or differentiation to their secretion of sEVs-particularly those under 200 nm-that mediate regenerative and immunomodulatory functions. Transitioning from cell therapies to sEV-based therapies offers clinical advantages, including reduced challenges with cell viability, storage, and administration, and improved pharmacological predictability. However, manufacturing MSC-sEV products faces challenges in defining critical quality attributes (CQAs) for consistent identity and potency. Variability arises from differences in cell sources, culture conditions, enrichment techniques, and the inherent heterogeneity of MSCs. Even the use of immortalized clonal MSC lines may not fully eliminate variability, as factors such as developmental processes, epigenetic modifications, or genetic drift could lead to the re-emergence of heterogeneity. Establishing robust potency CQAs is further complicated by the complex, multimodal modes of action of MSC-sEV products, which involve diverse mechanisms impacting various cell types and processes. Traditional models of EV mediated signalling suggesting direct internalization of sEVs by target cells are increasingly challenged due to inefficient EV-uptake and the high therapeutic efficacy observed. Instead, the Extracellular Modulation of Cells by EVs (EMCEV) model proposes that MSC-sEVs exert their effects by modulating the extracellular environment, enabling a "one EV to many cells" interaction. In conclusion, while MSC-sEV products hold significant therapeutic promise due to their multimodal action and functional redundancy, manufacturing challenges and the complexity of defining potency CQAs remain hurdles to clinical translation. A pragmatic approach focusing on identifying key potency-related CQAs based on specific mechanisms of action-while recognizing that "the process defines the product"-may facilitate the advancement of MSC-sEV therapeutics into clinical applications.
Collapse
Affiliation(s)
- Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | - Sai Kiang Lim
- Paracrine Therapeutics Pte. Ltd., Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
2
|
Badralmaa Y, Natarajan V. Aberrant Wnt/β-catenin signaling in the mesenchymal stem cells of LZTFL1-depleted mice leads to increased adipogenesis, with implications for obesity. J Biol Chem 2025; 301:108057. [PMID: 39662832 PMCID: PMC11770550 DOI: 10.1016/j.jbc.2024.108057] [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: 03/26/2024] [Revised: 11/05/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
Obesity is one of the main clinical characteristics associated with the heterogeneous genetic disorder Bardet-Biedl syndrome (BBS). Leucine zipper transcription factor like 1 (LZTFL1) is a member of the BBS gene family. Our work showed that Lztfl1knockout (LZKO) mice display the obesity phenotype as early as 3 months of age. Mesenchymal stem cells (MSCs) are multipotent stem cells that can differentiate into various cell types, including adipocytes. To understand the role of LZTFL1 in adipogenesis, we analyzed MSCs isolated from LZKO mouse compact bones (CB-MSCs). Compared to wildtype (WT), LZKO CB-MSCs had elongated primary cilia with tapered tips and increased levels of peroxisome proliferator-activated receptor γ (PPARγ), a key transcription factor that favors adipogenesis, and nuclear glucocorticoid receptor (GR), a transcription factor involved in Pparg activation. Also, LZKO CB-MSCs had a lower level of total β-catenin, a core factor of the antiadipogenic canonical Wnt/b-catenin signaling pathway involved in limiting the nuclear localization of GR. Interaction between caveolin1 (CAV1) and LRP6, the main receptor for canonical Wnt signaling, is known to be critical for Wnt pathway activation and β-catenin stabilization. Compared to WT cells, LZKO cells had elevated total, cell-surface, and lipid-raft-associated LRP6 and reduced CAV1, strongly indicating alterations in the components of the Wnt-signaling pathway. We show that in the absence of LZTFL1, adipogenesis-restraining Wnt/β-catenin signaling is inhibited, and adipogenesis-favorable factors are stimulated in CB-MSCs, leading to enhanced adipogenesis. Evidence provided here could help in understanding the mechanism and molecular basis of obesity in LZTFL1-defective patients.
Collapse
Affiliation(s)
- Yunden Badralmaa
- Laboratory of Molecular Cell Biology, Leidos Biomedical Research Inc, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Ven Natarajan
- Laboratory of Molecular Cell Biology, Leidos Biomedical Research Inc, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
| |
Collapse
|
3
|
Ahmad N, Anker A, Klein S, Dean J, Knoedler L, Remy K, Pagani A, Kempa S, Terhaag A, Prantl L. Autologous Fat Grafting-A Panacea for Scar Tissue Therapy? Cells 2024; 13:1384. [PMID: 39195271 PMCID: PMC11352477 DOI: 10.3390/cells13161384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024] Open
Abstract
Scars may represent more than a cosmetic concern for patients; they may impose functional limitations and are frequently associated with the sensation of itching or pain, thus impacting both psychological and physical well-being. From an aesthetic perspective, scars display variances in color, thickness, texture, contour, and their homogeneity, while the functional aspect encompasses considerations of functionality, pliability, and sensory perception. Scars located in critical anatomic areas have the potential to induce profound impairments, including contracture-related mobility restrictions, thereby significantly impacting daily functioning and the quality of life. Conventional approaches to scar management may suffice to a certain extent, yet there are cases where tailored interventions are warranted. Autologous fat grafting emerges as a promising therapeutic avenue in such instances. Fundamental mechanisms underlying scar formation include chronic inflammation, fibrogenesis and dysregulated wound healing, among other contributing factors. These mechanisms can potentially be alleviated through the application of adipose-derived stem cells, which represent the principal cellular component utilized in the process of lipofilling. Adipose-derived stem cells possess the capacity to secrete proangiogenic factors such as fibroblast growth factor, vascular endothelial growth factor and hepatocyte growth factor, as well as neurotrophic factors, such as brain-derived neurotrophic factors. Moreover, they exhibit multipotency, remodel the extracellular matrix, act in a paracrine manner, and exert immunomodulatory effects through cytokine secretion. These molecular processes contribute to neoangiogenesis, the alleviation of chronic inflammation, and the promotion of a conducive milieu for wound healing. Beyond the obvious benefit in restoring volume, the adipose-derived stem cells and their regenerative capacities facilitate a reduction in pain, pruritus, and fibrosis. This review elucidates the regenerative potential of autologous fat grafting and its beneficial and promising effects on both functional and aesthetic outcomes when applied to scar tissue.
Collapse
Affiliation(s)
- Nura Ahmad
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| | - Alexandra Anker
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| | - Silvan Klein
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| | - Jillian Dean
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Leonard Knoedler
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| | - Katya Remy
- Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Andrea Pagani
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| | - Sally Kempa
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| | - Amraj Terhaag
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| | - Lukas Prantl
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz–Josef–Strauß Allee 11, 93053 Regensburg, Germany; (A.A.); (S.K.); (L.K.); (A.P.); (S.K.); (A.T.); (L.P.)
| |
Collapse
|
4
|
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
|
5
|
Mundluru VK, Naidu MJ, Mundluru RT, Jeyaraman N, Muthu S, Ramasubramanian S, Jeyaraman M. Non-enzymatic methods for isolation of stromal vascular fraction and adipose-derived stem cells: A systematic review. World J Methodol 2024; 14:94562. [PMID: 38983657 PMCID: PMC11229868 DOI: 10.5662/wjm.v14.i2.94562] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/03/2024] [Accepted: 05/30/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Adipose-derived stem cells (ADSCs) and the stromal vascular fraction (SVF) have garnered substantial interest in regenerative medicine due to their potential to treat a wide range of conditions. Traditional enzymatic methods for isolating these cells face challenges such as high costs, lengthy processing time, and regu-latory complexities. AIM This systematic review aimed to assess the efficacy and practicality of non-enzymatic, mechanical methods for isolating SVF and ADSCs, comparing these to conventional enzymatic approaches. METHODS Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, a comprehensive literature search was conducted across multiple databases. Studies were selected based on inclusion criteria focused on non-enzymatic isolation methods for SVF and ADSCs from adipose tissue. The risk of bias was assessed, and a qualitative synthesis of findings was performed due to the methodological heterogeneity of the included studies. RESULTS Nineteen studies met the inclusion criteria, highlighting various mechanical techniques such as centrifugation, vortexing, and ultrasonic cavitation. The review identified significant variability in cell yield and viability, and the integrity of isolated cells across different non-enzymatic methods compared to enzymatic procedures. Despite some advantages of mechanical methods, including reduced processing time and avoidance of enzymatic reagents, the evidence suggests a need for optimization to match the cell quality and therapeutic efficacy achievable with enzymatic isolation. CONCLUSION Non-enzymatic, mechanical methods offer a promising alternative to enzymatic isolation of SVF and ADSCs, potentially simplifying the isolation process and reducing regulatory hurdles. However, further research is necessary to standardize these techniques and ensure consistent, high-quality cell yields for clinical applications. The development of efficient, safe, and reproducible non-enzymatic isolation methods could significantly advance the field of regenerative medicine.
Collapse
Affiliation(s)
- Vamsi Krishna Mundluru
- Department of Orthopaedics, MJ Naidu Super Speciality Hospital, Vijayawada 520002, Andhra Pradesh, India
- Department of Regenerative Medicine, StemC Clinics, Vijayawada 520002, Andhra Pradesh, India
| | - MJ Naidu
- Department of Orthopaedics, MJ Naidu Super Speciality Hospital, Vijayawada 520002, Andhra Pradesh, India
- Department of Regenerative Medicine, StemC Clinics, Vijayawada 520002, Andhra Pradesh, India
| | - Ravi Teja Mundluru
- Department of Orthopaedics, MJ Naidu Super Speciality Hospital, Vijayawada 520002, Andhra Pradesh, India
- Department of Regenerative Medicine, StemC Clinics, Vijayawada 520002, Andhra Pradesh, India
| | - Naveen Jeyaraman
- Department of Regenerative Medicine, StemC Clinics, Vijayawada 520002, Andhra Pradesh, India
- Department of Orthopaedics, ACS Medical College and Hospital, Dr. MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
| | - Sathish Muthu
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Department of Orthopaedics, Government Medical College and Hospital, Karur 639004, Tamil Nadu, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| | - Swaminathan Ramasubramanian
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Chennai 600002, Tamil Nadu, India
| | - Madhan Jeyaraman
- Department of Regenerative Medicine, StemC Clinics, Vijayawada 520002, Andhra Pradesh, India
- Department of Orthopaedics, ACS Medical College and Hospital, Dr. MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
| |
Collapse
|
6
|
Abraham M, Kori I, Vishwakarma U, Goel S. Comprehensive assessment of goat adipose tissue-derived mesenchymal stem cells cultured in different media. Sci Rep 2024; 14:8380. [PMID: 38600175 PMCID: PMC11006890 DOI: 10.1038/s41598-024-58465-1] [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: 07/12/2023] [Accepted: 03/29/2024] [Indexed: 04/12/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have demonstrated potential in treating livestock diseases that are unresponsive to conventional therapies. MSCs derived from goats, a valuable model for studying orthopaedic disorders in humans, offer insights into bone formation and regeneration. Adipose tissue-derived MSCs (ADSCs) are easily accessible and have a high capacity for expansion. Although the choice of culture media significantly influences the biological properties of MSCs, the optimal media for goat ADSCs (gADSCs) remains unclear. This study aimed to assess the effects of four commonly used culture media on gADSCs' culture characteristics, stem cell-specific immunophenotype, and differentiation. Results showed that MEM, DMEM/F12, and DMEM-LG were superior in maintaining cell morphology and culture parameters of gADSCs, such as cell adherence, metabolic activity, colony-forming potential, and population doubling. Conversely, DMEM-HG exhibited poor performance across all evaluated parameters. The gADSCs cultured in DMEM/F12 showed enhanced early proliferation and lower apoptosis. The cell surface marker distribution exhibited superior characteristics in gADSCs cultured in MEM and DMEM/F12. In contrast, the distribution was inferior in gADSCs cultured in DMEM-LG. DMEM/F12 and DMEM-LG culture media demonstrated a significantly higher potential for chondrogenic differentiation and DMEM-LG for osteogenic differentiation. In conclusion, DMEM/F12 is a suitable culture medium for propagating gADSCs as it effectively maintains cell morphology, growth parameters, proliferation and lower apoptosis while exhibiting desirable expression patterns of MSC-specific markers. These findings contribute to optimising culture conditions for gADSCs, enhancing their potential applications in disease treatment and regenerative medicine.
Collapse
Affiliation(s)
- Michelle Abraham
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
| | - Ibraz Kori
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- DBT-Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India
| | - Utkarsha Vishwakarma
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- DBT-Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India
| | - Sandeep Goel
- DBT-National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India.
- DBT-Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India.
| |
Collapse
|
7
|
Vazirzadeh M, Azarpira N, Vosough M, Ghaedi K. Galactosylation of rat natural scaffold for MSC differentiation into hepatocyte-like cells: A comparative analysis of 2D vs. 3D cell culture techniques. Biochem Biophys Rep 2023; 35:101503. [PMID: 37601454 PMCID: PMC10439353 DOI: 10.1016/j.bbrep.2023.101503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/05/2023] [Accepted: 06/21/2023] [Indexed: 08/22/2023] Open
Abstract
The liver plays a crucial role in drug detoxification, and the main source of liver transplants is brain-dead patients. However, the demand for transplants exceeds the available supply, leading to controversies in selecting suitable candidates for acute liver diseases. This research aimed to differentiate mesenchymal stem cells (MSCs) into hepatocyte-like cells using galactosylated rat natural scaffolds and comparing 2-D and 3-D cell culture methods. The study involved isolating and culturing Wharton's jelly cells from the umbilical cord, examining surface markers and adipogenic differentiation potential of MSCs, and culturing mesenchymal cells on galactosylated scaffolds. The growth and proliferation of stem cells on the scaffolds were evaluated using the MTT test, and urea synthesis was measured in different culture environments. Changes in gene expression were analyzed using real-time PCR. Flow cytometry results confirmed the presence of specific surface antigens on MSCs, indicating their identity, while the absence of a specific antigen indicated their differentiation into adipocytes. The MTT test revealed higher cell attachment to galactosylated scaffolds compared to the control groups. Urea secretion was observed in differentiated cells, with the highest levels in cells cultured on galactosylated scaffolds. Gene expression analysis showed differential expression patterns for OCT-4, HNF1, ALB, AFP, and CYP genes under different conditions. The findings indicated that hepatocyte-like cells derived from 3D cultures on galactosylated scaffolds exhibited superior characteristics compared to cells in other culture conditions. These cells demonstrated enhanced proliferation, stability, and urea secretion ability. The study also supported the differentiation potential of MSCs derived from Wharton's jelly umbilical cord into liver-like cells.
Collapse
Affiliation(s)
- Masoud Vazirzadeh
- Department of Cell and Molecular Biology and Microbiology, University of Isfahan, Isfahan, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
8
|
Valsecchi C, Croce S, Lenta E, Acquafredda G, Comoli P, Avanzini MA. TITLE: New therapeutic approaches in pediatric diseases: Mesenchymal stromal cell and mesenchymal stromal cell-derived extracellular vesicles as new drugs. Pharmacol Res 2023; 192:106796. [PMID: 37207738 DOI: 10.1016/j.phrs.2023.106796] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023]
Abstract
Mesenchymal Stromal Cell (MSC) clinical applications have been widely reported and their therapeutic potential has been documented in several diseases. MSCs can be isolated from several human tissues and easily expanded in vitro, they are able to differentiate in a variety of cell lineages, and they are known to interact with most immunological cells, showing immunosuppressive and tissue repair properties. Their therapeutic efficacy is closely associated with the release of bioactive molecules, namely Extracellular Vesicles (EVs), effective as their parental cells. EVs isolated from MSCs act by fusing with target cell membrane and releasing their content, showing a great potential for the treatment of injured tissues and organs, and for the modulation of the host immune system. EV-based therapies provide, as major advantages, the possibility to cross the epithelium and blood barrier and their activity is not influenced by the surrounding environment. In the present review, we deal with pre-clinical reports and clinical trials to provide data in support of MSC and EV clinical efficacy with particular focus on neonatal and pediatric diseases. Considering pre-clinical and clinical data so far available, it is likely that cell-based and cell-free therapies could become an important therapeutic approach for the treatment of several pediatric diseases.
Collapse
Affiliation(s)
- Chiara Valsecchi
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Stefania Croce
- Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Elisa Lenta
- Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Gloria Acquafredda
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Patrizia Comoli
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Maria Antonietta Avanzini
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| |
Collapse
|
9
|
Abd-Alameer M, Rajabibazl M, Esmaeilizadeh Z, Fazeli Z. SAG-dihydrochloride enhanced the expression of germ cell markers in the human bone marrow- mesenchymal stem cells (BM-MSCs) through the activation of GLI-independent hedgehog signaling pathway. Gene X 2023; 849:146902. [DOI: 10.1016/j.gene.2022.146902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 11/15/2022] Open
|
10
|
ÖZGENÇ Ö, ÖZEN A. Osteogenic Differentiation of Canine Adipose Derived Mesenchymal Stem Cells on B-TCP and B-TCP/Collagen Biomaterials. ANKARA ÜNIVERSITESI VETERINER FAKÜLTESI DERGISI 2022. [DOI: 10.33988/auvfd.1130705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Mesenchymal stem cells are adult stem cells that have the ability to differentiate into osteogenic, chondrogenic, adipogenic and myogenic lineages. In the field of orthopedics and traumatology, mesenchymal stem cells in combination with biomaterials are used especially for the treatment of bone fractures and diseases in both humans and animals. The purpose of this study is to promote growth, proliferation and osteogenic differentiation of mesenchymal stem cells that were isolated from the adipose tissue of canines on B-TCP (Beta-tricalcium phosphate) and B-TCP/Collagen biomaterials. MTT analysis was performed to test the cell adhesion and proliferation on B-TCP and B-TCP/Collagen biomaterials that were used to mimic the extracellular matrix of three-dimensional bone tissue. Scanning electron microscope analysis was performed to show general surface characters of B-TCP and B-TCP /Collagen biomaterials. The osteoinductive capacities of the B-TCP and B-TCP/Collagen biomaterials were determined by alkaline phosphatase and Von Kossa stainings, and RT-PCR analysis. The ALP activity of the B-TCP/Col containing material was significantly higher than the B-TCP on the first days. In terms of gene expression, there were no significant differences except 14th-day SPARC gene expression. The results of Von Kossa staining indicate that B-TCP/Col has above the desired level degradation capacity. As a result of this research, although it is advantageous in terms of alkaline phosphatase activity and osteogenic gene expression compared to B-TCP material, it is thought that B-TCP/Collagen biomaterial should be developed for use in bone tissue engineering due to its high degradation property.
Collapse
|
11
|
Shevela EY, Glebova TR, Kotova MA, Nitsa NA, Kozhevnikov YA, Meledina IV, Ostanin AA, Chernykh ER. Comparative Efficacy of the Stromal-Vascular Fraction Cells of Lipoaspirate and Hyaluronic Acid in the Treatment of Gonarthrosis: Results of an Interim Analysis. Bull Exp Biol Med 2022; 174:131-136. [PMID: 36437323 DOI: 10.1007/s10517-022-05661-6] [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: 02/21/2022] [Indexed: 11/29/2022]
Abstract
The use of cell technologies, in particular the stromal-vascular fraction of adipose tissue, is a new direction in the treatment of osteoarthritis of the weight-bearing joints. Stromal-vascular fraction cells have anti-inflammatory and immunomodulatory effects and are able to differentiate into connective tissue cells, including cartilage, tendons, and ligaments. Our clinical study showed the safety and good tolerability of intra-articular administration of autologous stromal-vascular fraction cells in 16 patients with severe manifestations of osteoarthritis. Single administration of stromal-vascular fraction cells led to more pronounced and stable (up to 12 months) clinical improvement in the main symptoms of the disease, including pain and functional activity of the affected joints, in comparison with intra-articular injection of hyaluronic acid (10 patients of the comparison group).
Collapse
Affiliation(s)
- E Ya Shevela
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia.
| | - T R Glebova
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - M A Kotova
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - N A Nitsa
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Yu A Kozhevnikov
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - I V Meledina
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - A A Ostanin
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - E R Chernykh
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| |
Collapse
|
12
|
Tambrchi P, Mahdavi AH, DaliriJoupari M, Soltani L. Polycaprolactone-co-polylactic acid nanofiber scaffold in combination with 5-azacytidine and transforming growth factor-β to induce cardiomyocyte differentiation of adipose-derived mesenchymal stem cells. Cell Biochem Funct 2022; 40:668-682. [PMID: 35924670 DOI: 10.1002/cbf.3728] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/11/2022]
Abstract
Adipose-derived mesenchymal stem cells (Ad-MSCs) are promising candidates for cardiac repair/regeneration. The application of copolymer nanoscaffolds has received great attention in tissue engineering to support differentiation and functional tissue organization toward effective tissue regeneration. The objective of the current study was to develop functional and bioactive scaffolds by combining polycaprolactone (PCL) and polylactic acid (PLA) for cardiomyocyte differentiation of human Ad-MSC (hAd-MSCs) in the absence or presence of 5-azacytidine and transforming growth factor-β (TGF-β). To that end, the human MSCs were extracted from human adipose tissue (AD). The cardiomyocyte differentiation potency of hAd-MSCs was evaluated on the novel synthetic PCL/PLA nanofiber scaffolds prepared in the absence and presence of 5-azacytidine and TGF-β supplements. A PCL/PLA nanofibrous scaffold was fabricated using the electrospinning method and its nanotopography and porous structure were characterized using scanning electron microscopy. In addition, the attachment of hAd-MSCs on the PCL/PLA scaffolds was semiquantitatively investigated. Compared with other treatments, the PCL/PLA nanofibrous scaffold supplemented with both 5-azacytidine and TGF-β was observed to differentiate hAd-MSCs into cardiomyocytes at Day 21 as evidenced by real-time PCR for cardiac-specific genes including cardiac troponin I (cTnI), GATA4, MYH7, and NKX2.5. In addition, flow cytometric analysis of cTnI-positive cells demonstrated that the cardiomyocyte differentiation of hAd-MSCs was more efficient on the PCL/PLA nanofibrous scaffold supplemented with both 5-azacytidine and TGF-β than it was in the other treatment groups. Generally speaking, the results show that PCL/PLA nanofibrous scaffolds may be applied as a platform for efficient differentiation of hAd-MSCs into functional cardiomyocytes.
Collapse
Affiliation(s)
- Parastoo Tambrchi
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Amir Hossein Mahdavi
- Department of Animal Science, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Morteza DaliriJoupari
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Leila Soltani
- Department of Animal Sciences, College of Agriculture and Natural Resources, Razi University, Kermanshah, Iran
| |
Collapse
|
13
|
Gadelkarim M, elmegeed AA, Hafez A, Awad AK, Shehata MA, AbouEl-Enein A, Alsadek ME, Deeb MA, Afifi AM. Safety and Efficacy of Adipose-Derived Mesenchymal Stem Cells for Knee Osteoarthritis: A Systematic Review and Meta-Analysis. Joint Bone Spine 2022; 89:105404. [DOI: 10.1016/j.jbspin.2022.105404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/18/2022] [Accepted: 04/28/2022] [Indexed: 11/27/2022]
|
14
|
Wang J, Hao R, Jiang T, Guo X, Zhou F, Cao L, Gao F, Wang G, Wang J, Ning K, Zhong C, Chen X, Huang Y, Xu J, Gao S. Rebuilding hippocampus neural circuit with hADSC-derived neuron cells for treating ischemic stroke. Cell Biosci 2022; 12:40. [PMID: 35379347 PMCID: PMC8981707 DOI: 10.1186/s13578-022-00774-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 03/14/2022] [Indexed: 12/17/2022] Open
Abstract
Background Human adipose-derived stem cells (hADSCs) have been demonstrated to be a promising autologous stem cell source for treating various neuronal diseases. Our study indicated that hADSCs could be induced into neuron-like cells in a stepwise manner that are characterized by the positive expression of MAP2, SYNAPSIN 1/2, NF-200, and vGLUT and electrophysiological activity. We first primed hADSCs into neuron-like cells (hADSC-NCs) and then intracerebrally transplanted them into MCAO reperfusion mice to further explore their in vivo survival, migration, integration, fate commitment and involvement in neural circuit rebuilding. Results The hADSC-NCs survived well and transformed into MAP2-positive, Iba1- or GFAP-negative cells in vivo while maintaining some proliferative ability, indicated by positive Ki67 staining after 4 weeks. hADSC-NCs could migrate to multiple brain regions, including the cortex, hippocampus, striatum, and hypothalamus, and further differentiate into mature neurons, as confirmed by action potential elicitation and postsynaptic currents. With the aid of a cell suicide system, hADSC-NCs were proven to have functionally integrated into the hippocampal memory circuit, where they contributed to spatial learning and memory rescue, as indicated by LTP improvement and subsequent GCV-induced relapse. In addition to infarction size shrinkage and movement improvement, MCAO-reperfused mice showed bidirectional immune modulation, including inhibition of the local proinflammatory factors IL-1α, IL-1β, IL-2, MIP-1β and promotion proinflammatory IP-10, MCP-1, and enhancement of the anti-inflammatory factors IL-15. Conclusion Overall, hADSC-NCs used as an intermediate autologous cell source for treating stroke can rebuild hippocampus neuronal circuits through cell replacement. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00774-x.
Collapse
Affiliation(s)
- Jian Wang
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Rui Hao
- Center of Translational Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200092, China.,Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai,, 200032,, China
| | - Tianfang Jiang
- Department of Neurology, Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, China
| | - Xuanxuan Guo
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Fei Zhou
- Department of Neurology, Third Affiliated Hospital of Navy Military Medical University, Shanghai, 200438, China
| | - Limei Cao
- Department of Neurology, Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, China
| | - Fengjuan Gao
- Zhoupu Hospital, Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Guangming Wang
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Juan Wang
- Department of Biotechnology and Molecular, Binzhou Medical College, Yantai, 264003, Shandong, People's Republic of China
| | - Ke Ning
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ, UK
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Xu Chen
- Department of Neurology, Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, China.
| | - Ying Huang
- Center of Translational Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200092, China.
| | - Jun Xu
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Shane Gao
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
15
|
Dai LG, Huang NC, Kang LY, Fu KY, Hsieh PS, Dai NT. An In Vitro Study of the Effects of Mechanical and Enzymatic Isolation of Stromal Vascular Fraction on Wound Healing. Ann Plast Surg 2022; 88:S13-S21. [PMID: 35225844 DOI: 10.1097/sap.0000000000003087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT The adipose-derived stromal vascular fraction (SVF) is considered to be an attractive source of stem cells in cell therapy. Besides stem cells, it also contains functional cells, such as macrophages, precursor cells, somatic stem cells, and pericytes. Collagenase digestion is the most frequently used method to isolate SVF, but it is time-consuming and costly and has some problems, such as infectious agents and immune reactions. In this research, we compared the yield, cell population ratios, and cell viability when isolating SVF by the ultrasonic physics (U-SVF) method and traditional enzymatic method (E-SVF). Then, we isolated exosomes from U-SVF and E-SVF, respectively, and cocultured them with fibroblasts to investigate the potential of applying this cell secretion in wound repair. The results showed that there was no significant difference between the ultrasonic method and enzymatic method in terms of cell viability, cell numbers, or the expression of CD markers of stem cells. However, exosome analysis identified a greater number and smaller size of exosome particles obtained by U-SVF. In terms of cell proliferation efficiency, although the proliferation efficiency of U-SVF was lower than that of E-SVF. Trilineage differentiation experiments revealed that both E-SVF and U-SVF had good differentiation ability, owing to high stem cell content. Finally, E-SVF and U-SVF exosomes were cocultured with fibroblasts. The efficiency of fibroblast migration increased in the SVF exosome treated groups, and the expression of related genes (integrin α5β1) was slightly upregulated; however, the expression of FAK, AKT, ERK, and RhoA was significantly upregulated at 24 hours. From the abovementioned experiments, we found that there was no significant difference in stem cell-related characteristics between SVF isolated by ultrasonic cavitation and SVF isolated by the enzymatic method. In addition, exosomes secreted by SVF may have excellent therapeutic effect on skin injuries, which provides a new viewpoint and therapeutic strategy for soft tissue repair.
Collapse
Affiliation(s)
- Lien-Guo Dai
- From the Department of Orthopaedic Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei
| | - Nien-Chi Huang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Lan-Ya Kang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Keng-Yen Fu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Pai-Shan Hsieh
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Niann-Tzyy Dai
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| |
Collapse
|
16
|
Wu H, Peng Z, Xu Y, Sheng Z, Liu Y, Liao Y, Wang Y, Wen Y, Yi J, Xie C, Chen X, Hu J, Yan B, Wang H, Yao X, Fu W, Ouyang H. Engineered adipose-derived stem cells with IGF-1-modified mRNA ameliorates osteoarthritis development. Stem Cell Res Ther 2022; 13:19. [PMID: 35033199 PMCID: PMC8760691 DOI: 10.1186/s13287-021-02695-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/20/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA), a prevalent degenerative disease characterized by degradation of extracellular matrix (ECM), still lacks effective disease-modifying therapy. Mesenchymal stem cells (MSCs) transplantation has been regarded as the most promising approach for OA treatment while engrafting cells alone might not be adequate for effective regeneration. Genetic modification has been used to optimize MSC-based therapy; however, there are still significant limitations that prevent the clinical translation of this therapy including low efficacy and safety concerns. Recently, chemically modified mRNA (modRNA) represents a promising alternative for the gene-enhanced MSC therapy. In this regard, we hypothesized that adipose derived stem cells (ADSCs) engineered with modRNA encoding insulin-like growth factor 1 (IGF-1) were superior to native ADSCs on ameliorating OA development. METHODS Mouse ADSCs were acquired from adipose tissue and transfected with modRNAs. First, the kinetics and efficacy of modRNA-mediated gene transfer in mouse ADSCs were analyzed in vitro. Next, we applied an indirect co-culture system to analyze the pro-anabolic potential of IGF-1 modRNA engineered ADSCs (named as IGF-1-ADSCs) on chondrocytes. Finally, we evaluated the cell retention and chondroprotective effect of IGF-1-ADSCs in vivo using fluorescent labeling, histology and immunohistochemistry. RESULTS modRNA transfected mouse ADSCs with high efficiency (85 ± 5%) and the IGF-1 modRNA-transfected ADSCs facilitated burst-like production of bio-functional IGF-1 protein. In vitro, IGF-1-ADSCs induced increased anabolic markers expression of chondrocytes in inflammation environment compared to untreated ADSCs. In a murine OA model, histological and immunohistochemical analysis of knee joints harvested at 4 weeks and 8 weeks after OA induction suggested IGF-1-ADSCs had superior therapeutic effect over native ADSCs demonstrated by lower histological OARSI score and decreased loss of cartilage ECM. CONCLUSIONS These findings collectively supported the therapeutic potential of IGF-1-ADSCs for clinical OA management and cartilage repair.
Collapse
Affiliation(s)
- Haoyu Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi Peng
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Xu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Zixuan Sheng
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanshan Liu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Youguo Liao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yin Wang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Ya Wen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Junzhi Yi
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Chang Xie
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuri Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiajie Hu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Bingqian Yan
- Institute of Pediatric Translational Medicine, Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 310003, China
| | - Huijing Wang
- Institute of Pediatric Translational Medicine, Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 310003, China
| | - Xudong Yao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Fu
- Institute of Pediatric Translational Medicine, Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 310003, China.
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China. .,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China. .,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China. .,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| |
Collapse
|
17
|
Abstract
Exosomes are extracellular vesicles (EVs) released from cells that are a part of many biological and pathological processes, especially in intercellular communication. These vesicles are involved cell signaling, influence tissue and immune response, and serve as biomarkers for diseases. Most interesting are the exosomes that are released from mesenchymal stem cells (MSCs) for inflammation in joint diseases. Preliminary studies have demonstrated the advantages of using EVs rather than MSCs for cell free therapy. Research on exosomes have shown promising results as biomarkers for tracking the pathogenesis and prognosis of inflammatory arthritis. Therapeutically, animal studies have demonstrated immunosuppression, reversing inflammation, increasing chondrocyte proliferation, and drug delivery properties. The field of exosomes continues to develop and more basic science and clinical studies with safety and efficacy studies are needed.
Collapse
Affiliation(s)
- William Fang
- Department of Orthopaedic Surgery, Keck
School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - C. Thomas Vangsness
- Department of Orthopaedic Surgery, Keck
School of Medicine, University of Southern California, Los Angeles, CA, USA,C. Thomas Vangsness Jr., Department of
Orthopaedic Surgery, Keck School of Medicine, University of Southern California,
1520 San Pablo Street 2000, Los Angeles, CA 90033, USA.
| |
Collapse
|
18
|
Wang F, Yang G, Xiao Y, He C, Cai G, Song E, Li Y. Effects of Tissue-engineered Bone by Coculture of Adipose-derived Stem Cells and Vascular Endothelial Cells on Host Immune Status. Ann Plast Surg 2021; 87:689-693. [PMID: 34818288 DOI: 10.1097/sap.0000000000002824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
AIM The study aimed to explore the effects of tissue-engineered bone constructed with partially deproteinized biologic bone (PDPBB) and coculture of adipose-derived stem cells (ADSCs) and vascular endothelial cells (VECs) on host immune status, providing a very useful clue for the future development of bone engineering. METHODS Tissue-engineered bones constructed by PDPBB and ADSCs, VECs or coculture of them were implanted into the muscle bag of bilateral femurs of Sprague-Dawley rats. Partially deproteinized biologic bone alone and blank control were also implanted. After transplantation, the proliferation of implanted seed cells in tissue-engineered bones was labeled by bromodeoxyuridine staining. Moreover, the changes of T-lymphocyte subpopulations, including CD3 + CD4+ and CD3 + CD8+ in peripheral blood were then detected using flow cytometry to analyze the immune rejection of tissue-engineered bone implantation based on peripheral blood CD4/CD8 ratios. RESULTS After transplantation, the proliferation of implanted seed cells was observed in tissue-engineered bones of different groups. At different time points after transplantation, the CD4+/CD8+ ratio in peripheral blood of PDPBB + ADSCs, PDPBB + coculture, and blank control groups did not exhibit significant change. Although the CD4+/CD8+ ratio in peripheral blood of PDPBB + VECs group was significantly higher than other group at 1 week after transplantation, that of PDPBB + VECs and PDPBB + coculture group was significantly decreased at 8 week after transplantation compared with that of blank control group. CONCLUSIONS Our results indicated that there was no significant immune rejection after transplantation of tissue-engineered bone constructed with PDPBB and coculture of ADSCs and VECs as seed cells.
Collapse
Affiliation(s)
- Fuke Wang
- From the Department of Sports Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | | | | | | | | | | | | |
Collapse
|
19
|
Luo S, Ai Y, Xiao S, Wang B, Wang Y. Functional hit 1 (FH1)-based rapid and efficient generation of functional hepatocytes from human mesenchymal stem cells: a novel strategy for hepatic differentiation. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1087. [PMID: 34422999 PMCID: PMC8339809 DOI: 10.21037/atm-21-2829] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 06/25/2021] [Indexed: 12/19/2022]
Abstract
Background Because the liver is central to the physiology of the body, primary hepatocytes are widely used in liver pathology and physiological research, such as liver drug screening, bioartificial liver support system, and cell therapy for liver diseases. However, the source of primary hepatocytes is limited. We describe a novel non-transgenic protocol that facilitates the rapid generation of hepatocyte-like cells from human umbilical cord-derived mesenchymal stem cells (hUC-MSCs), providing a new source of functional hepatocytes. Methods In this study, we used hUC-MSCs and human induced pluripotent cells (iPSCs) derived mesenchymal stem cells (iMSCs) to investigate the new induction strategy. Passage 3 MSCs were induced into hepatocyte-like cells using small-molecule compounds combined with cell factors in vitro. Functional hit 1 (FH1), a promising small molecule compound was achieved to replace HGF in the hepatocyte maturation stage to induce the hepatocyte-like cells differentiation. Results We rapidly induced hUC-MSCs and human iMSCs into hepatocyte-like cells within 10 days in vitro, and the cells were morphologically similarly to both hepatocytes derived from the hepatocyte growth factor (HGF)-based method and the primary hepatocytes. They expressed mature hepatocyte special genes and achieved functions such as glycogen storage, albumin expression, urea secretion, cytochrome P450 activity, Low-density lipoprotein (LDL) uptake, and indocyanine green (ICG) uptake. Conclusions We successfully established a small-molecule protocol without using HGF to differentiate MSCs into hepatocyte-like cells, which provides a rapid and cost-effective platform for in vitro studies of liver disease.
Collapse
Affiliation(s)
- Sang Luo
- State Key Laboratory of Virology, School of Life Sciences, Wuhan University, Wuhan, China
| | - Yang Ai
- State Key Laboratory of Virology, School of Life Sciences, Wuhan University, Wuhan, China
| | - Shuai Xiao
- State Key Laboratory of Virology, School of Life Sciences, Wuhan University, Wuhan, China
| | - Ben Wang
- State Key Laboratory of Virology, School of Life Sciences, Wuhan University, Wuhan, China
| | - Yefu Wang
- State Key Laboratory of Virology, School of Life Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
20
|
Mannino G, Russo C, Longo A, Anfuso CD, Lupo G, Lo Furno D, Giuffrida R, Giurdanella G. Potential therapeutic applications of mesenchymal stem cells for the treatment of eye diseases. World J Stem Cells 2021; 13:632-644. [PMID: 34249232 PMCID: PMC8246249 DOI: 10.4252/wjsc.v13.i6.632] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/07/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
Stem cell-based treatments have been extensively explored in the last few decades to develop therapeutic strategies aimed at providing effective alternatives for those human pathologies in which surgical or pharmacological therapies produce limited effects. Among stem cells of different sources, mesenchymal stem cells (MSCs) offer several advantages, such as the absence of ethical concerns, easy harvesting, low immunogenicity and reduced tumorigenesis risks. Other than a multipotent differentiation ability, MSCs can release extracellular vesicles conveying proteins, mRNA and microRNA. Thanks to these properties, new therapeutic approaches have been designed for the treatment of various pathologies, including ocular diseases. In this review, the use of different MSCs and different administration strategies are described for the treatment of diabetic retinopathy, glaucoma, and retinitis pigmentosa. In a large number of investigations, positive results have been obtained by in vitro experiments and by MSC administration in animal models. Most authors agree that beneficial effects are likely related to MSC paracrine activity. Based on these considerations, many clinical trials have already been carried out. Overall, although some adverse effects have been described, promising outcomes are reported. It can be assumed that in the near future, safer and more effective protocols will be developed for more numerous clinical applications to improve the quality of life of patients affected by eye diseases.
Collapse
Affiliation(s)
- Giuliana Mannino
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania 95123, Italy
| | - Cristina Russo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania 95123, Italy
| | - Anna Longo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania 95123, Italy
| | - Carmelina Daniela Anfuso
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania 95123, Italy
| | - Gabriella Lupo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania 95123, Italy
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania 95123, Italy.
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania 95123, Italy
| | - Giovanni Giurdanella
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania 95123, Italy
| |
Collapse
|
21
|
Rahmani-Moghadam E, Zarrin V, Mahmoodzadeh A, Owrang M, Talaei-Khozani T. Comparison of the Characteristics of Breast Milk-derived Stem Cells with the Stem Cells Derived from the Other Sources: A Comparative Review. Curr Stem Cell Res Ther 2021; 17:71-90. [PMID: 34161214 DOI: 10.2174/1574888x16666210622125309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/14/2021] [Accepted: 03/28/2021] [Indexed: 11/22/2022]
Abstract
Breast milk (BrM) not only supplies nutrition, but it also contains a diverse population of cells. It has been estimated that up to 6% of the cells in human milk possess the characteristics of mesenchymal stem cells (MSC). Available data also indicate that these cells are multipotent and capable of self-renewal and differentiation with other cells. In this review, we have compared different characteristics, such as CD markers, differentiation capacity, and morphology of stem cells, derived from human breast milk (hBr-MSC) with human bone marrow (hBMSC), Wharton's jelly (WJMSC), and human adipose tissue (hADMSC). Through the literature review, it was revealed that human breast milk-derived stem cells specifically express a group of cell surface markers, including CD14, CD31, CD45, and CD86. Importantly, a group of markers, CD13, CD29, CD44, CD105, CD106, CD146, and CD166, were identified, which were common in the four sources of stem cells. WJMSC, hBMSC, hADMSC, and hBr-MSC are potently able to differentiate into the mesoderm, ectoderm, and endoderm cell lineages. The ability of hBr-MSCs todifferentiate into the neural stem cells, neurons, adipocyte, hepatocyte, chondrocyte, osteocyte, and cardiomyocytes has made these cells a promising source of stem cells in regenerative medicine, while isolation of stem cells from the commonly used sources, such as bone marrow, requires invasive procedures. Although autologous breast milk-derived stem cells are an accessible source for women who are in the lactation period, breast milk can be considered as a source of stem cells with high differentiation potential without any ethical concern.
Collapse
Affiliation(s)
- Ebrahim Rahmani-Moghadam
- Department of Anatomical sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahideh Zarrin
- Laboratory for Stem Cell Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Mahmoodzadeh
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Marzieh Owrang
- Department of Anatomical sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Talaei-Khozani
- Department of Anatomical sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
22
|
REIS BPZCD, ORGE ID, SAMPAIO GLDA, DALTRO SRT, SANTOS RRD, MEIRA CS, SOARES MBP. Mesenchymal Stem cells in the context of canine atopic dermatitis: A Review. REVISTA BRASILEIRA DE SAÚDE E PRODUÇÃO ANIMAL 2021. [DOI: 10.1590/s1519-99402122242021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
ABSTRACT Canine atopic dermatitis (CAD) is a chronic inflammatory skin disease and has a high frequency among dermatological diseases. The interaction of genetic factors, skin and environmental conditions affect the expression of the disease, developing a complex pathology. Current multimodal treatment has numerous adverse effects and variations in its efficacy and safety, demonstrating the need to develop safe and effective therapeutic resources for patients with CAD. Mesenchymal stem cells (MSCs) are multipotent cells, with special characteristics, such as self-renewal, immunomodulatory properties, and de-differentiation, making them useful for several clinical problems. The discovery of the immunosuppressive effect of MSCs on T cells has opened the potential for new perspectives with its use as a therapeutic agent for immune diseases, such as CAD. The scarce number of research using the MSC as a treatment for CAD result in the lack of knowledge about the benefits and possible protocols to be followed for the use of this cell therapy. In this review, we highlighted the clinical studies and potential biological mechanisms of MSC-based cell therapy effects attenuating canine atopic dermatitis compared to conventional treatment, which might lead to a safe improvement of the animal’s clinical condition in a short period without causing adverse effects.
Collapse
Affiliation(s)
| | | | | | | | | | - Cássio Santana MEIRA
- Oswaldo Cruz Foundation (FIOCRUZ), Brazil; University Center SENAI/CIMATEC, Brazil
| | | |
Collapse
|
23
|
Kim SH, Park YB. Editorial Commentary: Stem Cell Treatment in Knee Osteoarthritis: What for? Pain Management or Cartilage Regeneration? Arthroscopy 2021; 37:359-361. [PMID: 33384092 DOI: 10.1016/j.arthro.2020.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 02/02/2023]
Abstract
The efficacy of mesenchymal stem cells regarding clinical outcomes and cartilage regeneration in knee osteoarthritis remains unclear; however, their theoretical role in multilineage cellular differentiation and immunomodulation of the arthritic cascade has been investigated. Several studies have reported that the use of stem cell therapy for knee osteoarthritis helps in pain improvement, but its effect on cartilage regeneration has not yet been explored. Moreover, numerous studies have reported high heterogeneity in the cell sources, as well as methods of culture expansion or cell concentration, and differences in delivery methods, assessment tools, and concomitant surgical procedures, which could affect the clinical outcomes or evaluation of cartilage regeneration potency. Furthermore, future studies are warranted to examine these factors in detail to interpret the results of mesenchymal stem cell treatment for knee osteoarthritis.
Collapse
|
24
|
Dai W, Leng X, Wang J, Shi Z, Cheng J, Hu X, Ao Y. Intra-Articular Mesenchymal Stromal Cell Injections Are No Different From Placebo in the Treatment of Knee Osteoarthritis: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Arthroscopy 2021; 37:340-358. [PMID: 33098949 DOI: 10.1016/j.arthro.2020.10.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/08/2020] [Accepted: 10/08/2020] [Indexed: 02/02/2023]
Abstract
PURPOSE To evaluate the efficacy and safety of intra-articular mesenchymal stromal cells (MSCs) injections for knee osteoarthritis (OA) treatment. METHODS We performed a systematic literature search in PubMed, Embase, Scopus, and the Cochrane Library through April 2020 to identify level I randomized controlled trials (RCTs) that evaluated the clinical efficacy of MSCs versus control treatments for knee OA. Outcomes were analyzed on an intention-to-treat basis with random-effects models. RESULTS A total of 13 RCTs were included in the meta-analysis. Compared with placebo, there was no significant difference in VAS for pain (mean difference [MD] 1.62, 95% confidence interval [CI -0.60 to 3.85), WOMAC pain score (MD 1.88, 95% CI -0.21 to 3.98), WOMAC function score (MD -0.67, 95% CI -6.54 to 5.19), or WOMAC stiffness score (MD 0.64, 95% CI -0.86 to 2.14) for MSCs. Moreover, the smallest treatment effect of VAS for pain, WOMAC pain score, WOMAC function score, and WOMAC stiffness score did not exceed the minimum clinically important difference (MCID). Additionally, there was no significant difference in percentage of patients crossing the MCID threshold between MSC and placebo groups for VAS for pain (relative risk [RR] 0.93, 95% CI 0.55 to 1.57) or WOMAC total score (RR 0.40, 95% CI 0.13 to 1.21). Compared with hyaluronic acid (HA), MSC injection was associated with significantly better improvement in VAS for pain (MD 2.00, 95% CI 0.94 to 3.07), WOMAC pain score (MD 4.58, 95% CI 0.49 to 8.67), WOMAC total score (MD 14.86, 95% CI 10.59 to 19.13), and WOMAC stiffness score (MD 1.85, 95% CI 0.02 to 3.69). However, the smallest treatment effect of VAS for pain, WOMAC pain score, WOMAC function score, and WOMAC stiffness score did not exceed the MCID. Moreover, there was no significant difference in percentage of patients crossing the MCID threshold between MSC and HA groups for WOMAC total score (RR 0.57, 95% CI 0.21 to 1.55). We also found that MSCs did not increase adverse events compared with HA and placebo. CONCLUSIONS Intra-articular MSC injection was not found to be superior to placebo in pain relief and functional improvement for patients with symptomatic knee OA. However, additional direct testing and combination trials of different type of cells, doses, and number of injections of MSCs are required to further enhance clinical decision making for people with symptomatic knee OA. LEVEL OF EVIDENCE I, meta-analysis of level I studies.
Collapse
Affiliation(s)
- Wenli Dai
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, People's Republic of China
| | - Xi Leng
- Medical Imaging Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong, People's Republic of China
| | - Jian Wang
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangdong, People's Republic of China
| | - Zhanjun Shi
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangdong, People's Republic of China
| | - Jin Cheng
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, People's Republic of China
| | - Xiaoqing Hu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, People's Republic of China.
| | - Yingfang Ao
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, People's Republic of China.
| |
Collapse
|
25
|
Miller H, De Leo N, Badach J, Lin A, Williamson J, Bonawitz S, Ostrovsky O. Role of marijuana components on the regenerative ability of stem cells. Cell Biochem Funct 2020; 39:432-441. [PMID: 33349985 DOI: 10.1002/cbf.3609] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/01/2020] [Accepted: 12/13/2020] [Indexed: 12/17/2022]
Abstract
Stem cell therapy promotes tissue regeneration and wound healing. Efforts have been made to prime stem cells to enhance their regenerative abilities. Certain marijuana components, namely the non-psychoactive cannabidiol (CBD) and psychoactive tetrahydrocannabinol (THC), are defined as immunomodulators.9 We test whether two sources of stem cells, primed with CBD or THC, would demonstrate improved regenerative abilities. Human adipose-derived stem cells (ASCs) and bone marrow-derived stem cells (BMDSCs), not obtained from the same individual, were treated with low (300 nM) or high (3 μM) concentration CBD. Porcine ASCs and BMDSCs were isolated from a single pig, and treated with either low or high concentrations of CBD or THC. Transwell migration and MTT proliferation assays were performed on the human ASCs and BMDSCs. Also, transwell migration assay was performed on the porcine ASCs and BMDSCs. Finally, a wound healing scratch assay in porcine primary fibroblasts (PFs) was performed, co-cultured with the cannabinoid-treated ASCs. CBD priming at low concentration induces migration by 180% (P < .01) in porcine ASCs, and by only 93% (P < .02) in porcine BMDSCs. In porcine stem cells, THC priming at low concentration induces migration by 91.6% (P < .01) in ASCs but by only 44.3% (P < .03) in BMDSCs. Compared to PFs co-cultured with untreated ASCs, PFs co-cultured with low CBD-primed ASCs had 75% faster wound closure at 18 hours (P < .01). CBD and THC priming of ASCs and BMDSCs, particularly at lower doses, enhances a number of regenerative parameters, suggesting that these major marijuana components may improve stem cell-based therapies. SIGNIFICANCE OF THE STUDY: Our study demonstrates that cannabinoids can enhance the regenerative capacity of two major sources of stem cells, adipose- and bone marrow-derived, from human and porcine donors. Stem cell isolation and expansion is invasive, costly and time consuming. Stem cells with improved regenerative properties may be effective in the treatment of acute or chronic wounds. This is the first study to compare the priming potential of two sources of stem cells from the same animal, with the same genetic and epigenetic profile, as well as the first to prime with THC.
Collapse
Affiliation(s)
- Henry Miller
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| | - Nicholas De Leo
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| | - Jeremy Badach
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| | - Andrew Lin
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| | - John Williamson
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| | - Steven Bonawitz
- Department of Surgery, Cooper University Hospital, Camden, New Jersey, USA
| | - Olga Ostrovsky
- Cooper Research Institute, Cooper University Hospital, Camden, New Jersey, USA
| |
Collapse
|
26
|
Antioxidant and Biological Properties of Mesenchymal Cells Used for Therapy in Retinitis Pigmentosa. Antioxidants (Basel) 2020; 9:antiox9100983. [PMID: 33066211 PMCID: PMC7602011 DOI: 10.3390/antiox9100983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/04/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
Both tissue repair and regeneration are a priority in regenerative medicine. Retinitis pigmentosa (RP), a complex retinal disease characterized by the progressive loss of impaired photoreceptors, is currently lacking effective therapies: this represents one of the greatest challenges in the field of ophthalmological research. Although this inherited retinal dystrophy is still an incurable genetic disease, the oxidative damage is an important pathogenetic element that may represent a viable target of therapy. In this review, we summarize the current neuroscientific evidence regarding the effectiveness of cell therapies in RP, especially those based on mesenchymal cells, and we focus on their therapeutic action: limitation of both oxidative stress and apoptotic processes triggered by the disease and promotion of cell survival. Cell therapy could therefore represent a feasible therapeutic option in RP.
Collapse
|
27
|
|
28
|
Lee JS, Chae S, Yoon D, Yoon D, Chun W, Kim GH. Angiogenic factors secreted from human ASC spheroids entrapped in an alginate-based hierarchical structure via combined 3D printing/electrospinning system. Biofabrication 2020; 12:045028. [PMID: 32946427 DOI: 10.1088/1758-5090/abaf9a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Human adipose-derived stem cell spheroids have been widely used in the treatment or regeneration of damaged skin tissues, and their success is believed to be due in part to angiogenic factors released from the spheroids. To achieve the sustained release of bioactive components from implanted spheroids within a defective area, the use of a biocompatible scaffolding biomaterial is required. In this study, we developed an alginate-based scaffolding structure, which was processed using three-dimensional printing and electrospinning for use as a spheroid-entrapping structure. A micro-sized alginate strut and electrospun alginate nanofibers functioned not only to firmly entrap the spheroids, but also to enable the stable release of various angiogenic and wound healing-related factors. We also demonstrated the function of these factors using a tube-forming assay and found that conditioned media from the spheroid-scaffold group improved capillary-like structure formation in human umbilical vein endothelial cells compared to the single cell-scaffold group. Our results suggest that this spheroid-entrapping alginate hybrid structure could represent a new platform for stem cell therapy using spheroid transplantation.
Collapse
Affiliation(s)
- Ji-Seon Lee
- Burn Institute, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, Seoul, Republic of Korea. These authors contributed equally to this work
| | | | | | | | | | | |
Collapse
|
29
|
Limoli PG, Limoli CSS, Morales MU, Vingolo EM. Mesenchymal stem cell surgery, rescue and regeneration in retinitis pigmentosa: clinical and rehabilitative prognostic aspects. Restor Neurol Neurosci 2020; 38:223-237. [PMID: 32310198 PMCID: PMC7504992 DOI: 10.3233/rnn-190970] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Purpose: To assess whether treatment with the Limoli Retinal Restoration Technique (LRRT) can be performed in patients with retinitis pigmentosa (RP), grafting the autologous cells in a deep scleral pocket above the choroid of each eye to exert their beneficial effect on the residual retinal cells. Methods: The patients were subjected to a complete ophthalmological examination, including best corrected visual acuity (BCVA), close-up visus measurements, spectral domain-optical coherence tomography (SD-OCT), microperimetry (MY), and electroretinography (ERG). Furthermore, the complete ophthalmological examination was carried out at baseline (T0) and at 6 months (T180) after surgery. The Shapiro–Wilk test was used to assess the normality of distribution of the investigated parameters. A mixed linear regression model was used to analyse the difference in all the studied parameters at T0 and T180, and to compare the mean change between the two groups. All statistical analyses were performed with STATA 14.0 (Collage Station, Texas, USA). Results: LRRT treatment was performed in 34 eyes of 25 RP patients recruited for the study. The eyes were classified in two groups on the basis of foveal thickness (FT) assessed by SD-OCT: 14 eyes in Group A (FT≤190μm) and the remaining 20 ones in Group B (FT > 190μm). Although it had not reached the statistical significance, Group B showed a better improvement in BCVA, residual close-up visus and sensitivity than Group A. Conclusions: Previous studies have described the role of LRRT in slowing down retinal degenerative diseases. Consequently, this surgical procedure could improve the clinical and rehabilitative prognostic parameters in RP patients. On the other hand, further clinical research and studies with longer follow-up will be needed to evaluate its efficacy.
Collapse
Affiliation(s)
| | | | - Marco Ulises Morales
- Division of Clinical Neurosciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Enzo Maria Vingolo
- Department of Sense Organs, Faculty of Medicine and Odontology, Sapienza University of Rome, p.le A. Moro, Rome, Italy
| |
Collapse
|
30
|
Terraza-Aguirre C, Campos-Mora M, Elizondo-Vega R, Contreras-López RA, Luz-Crawford P, Jorgensen C, Djouad F. Mechanisms behind the Immunoregulatory Dialogue between Mesenchymal Stem Cells and Th17 Cells. Cells 2020; 9:cells9071660. [PMID: 32664207 PMCID: PMC7408034 DOI: 10.3390/cells9071660] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/03/2020] [Accepted: 07/05/2020] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) exhibit potent immunoregulatory abilities by interacting with cells of the adaptive and innate immune system. In vitro, MSCs inhibit the differentiation of T cells into T helper 17 (Th17) cells and repress their proliferation. In vivo, the administration of MSCs to treat various experimental inflammatory and autoimmune diseases, such as rheumatoid arthritis, type 1 diabetes, multiple sclerosis, systemic lupus erythematosus, and bowel disease showed promising therapeutic results. These therapeutic properties mediated by MSCs are associated with an attenuated immune response characterized by a reduced frequency of Th17 cells and the generation of regulatory T cells. In this manuscript, we review how MSC and Th17 cells interact, communicate, and exchange information through different ways such as cell-to-cell contact, secretion of soluble factors, and organelle transfer. Moreover, we discuss the consequences of this dynamic dialogue between MSC and Th17 well described by their phenotypic and functional plasticity.
Collapse
Affiliation(s)
- Claudia Terraza-Aguirre
- IRMB, University of Montpellier, INSERM, F-34090 Montpellier, France; (C.T.-A.); (R.A.C.-L.)
| | | | - Roberto Elizondo-Vega
- Facultad de Ciencias Biológicas, Departamento de Biología Celular, Laboratorio de Biología Celular, Universidad de Concepción, Concepción 4030000, Chile;
| | | | - Patricia Luz-Crawford
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago 7620001, Chile;
| | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, F-34090 Montpellier, France; (C.T.-A.); (R.A.C.-L.)
- CHU Montpellier, F-34295 Montpellier, France
- Correspondence: (C.J.); (F.D.); Tel.: +33-(0)-4-67-33-77-96 (C.J.); +33-(0)-4-67-33-04-75 (F.D.)
| | - Farida Djouad
- IRMB, University of Montpellier, INSERM, F-34090 Montpellier, France; (C.T.-A.); (R.A.C.-L.)
- Correspondence: (C.J.); (F.D.); Tel.: +33-(0)-4-67-33-77-96 (C.J.); +33-(0)-4-67-33-04-75 (F.D.)
| |
Collapse
|
31
|
He W, Zheng Y, Feng Q, Elkhooly TA, Liu X, Yang X, Wang Y, Xie Y. Silver nanoparticles stimulate osteogenesis of human mesenchymal stem cells through activation of autophagy. Nanomedicine (Lond) 2020; 15:337-353. [DOI: 10.2217/nnm-2019-0026] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: Previously, different results have been achieved regarding effects of silver nanoparticles (Ag NPs) on osteogenesis of stem cells and the mechanisms have not been disclosed yet, which are quite important for potential application of Ag NPs in bone reconstruction. Materials & methods: Effects of Ag NPs on osteogenesis of human mesenchymal stem cells (hMSCs) with underlying mechanisms were investigated. Results: Ag NPs at 2.5 and 5 μg/ml increased osteogenic proteins expression and mineralization of hMSCs. Meanwhile, autophagy was activated by Ag NPs and it could be inhibited by 3-methyladenine. Furthermore, osteogenesis induced by Ag NPs could also be reversed by 3-methyladenine. Conclusion: These findings suggest that autophagy is involved in stimulating osteogenesis of hMSCs induced by Ag NPs.
Collapse
Affiliation(s)
- Wei He
- School of Materials Science & Engineering, University of Science & Technology Beijing, Beijing, 100083, PR China
| | - Yudong Zheng
- School of Materials Science & Engineering, University of Science & Technology Beijing, Beijing, 100083, PR China
| | - Qingling Feng
- State Key Laboratory of New Ceramics & Fine Processing, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Tarek A Elkhooly
- Department of Refractories, Ceramics & Building Materials, National Research Centre, Dokki, 12622, Cairo, Egypt
| | - Xujie Liu
- State Key Laboratory of New Ceramics & Fine Processing, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Xing Yang
- State Key Laboratory of New Ceramics & Fine Processing, School of Materials Science & Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Yansen Wang
- School of Materials Science & Engineering, University of Science & Technology Beijing, Beijing, 100083, PR China
| | - Yajie Xie
- School of Materials Science & Engineering, University of Science & Technology Beijing, Beijing, 100083, PR China
| |
Collapse
|
32
|
Liu GB, Pan YM, Liu YS, Hu JH, Zhang XD, Zhang DW, Wang Y, Feng YK, Yu JB, Cheng YX. Ghrelin promotes neural differentiation of adipose tissue-derived mesenchymal stem cell via AKT/mTOR and β-catenin signaling pathways. Kaohsiung J Med Sci 2020; 36:405-416. [PMID: 32003536 DOI: 10.1002/kjm2.12188] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/07/2020] [Indexed: 02/04/2023] Open
Abstract
Adipose tissue-derived mesenchymal stem cells (ADSCs) are multipotent cells that can differentiate into various cell types. This study aimed to investigate the effect of ghrelin on the neural differentiation of rat ADSCs and underlying molecular mechanisms. Rat ADSCs were isolated and third-passage ADSCs were used in this study. The isolated ADSCs were characterized by flow cytometry analysis for MSCs' surface expression markers as evidenced by positive for CD90, CD44, and CD29 and negative for CD34, CD45, and CD11b/2f/c. The multilineage differentiation of ADSCs was confirmed by adipogenic, osteogenic, and neural differentiation. After induction of neurogenesis, the differentiated cells were identified by development of neuron-like morphology and expression of neural markers including glial fibrillary acidic protein, Nestin, MAP2, and β-Tubulin III using immunofluorescence and western blot. Ghrelin concentration dependently elevated the proportion of neural-like cells and branching dendrites, as well as upregulated the expression of neural markers. Further, the expression of nuclear β-catenin, p-GSK-3β, p-AKT, and p-mTOR was increased by ghrelin, indicating an activation of β-catenin and AKT/mTOR signaling after the ghrelin treatment. Importantly, inhibition of β-catenin or AKT/mTOR signaling suppressed ghrelin-induced neurogenesis. Therefore, we demonstrate that ghrelin promotes neural differentiation of ADSCs through the activation of β-catenin and AKT/mTOR signaling pathways.
Collapse
Affiliation(s)
- Gui-Bo Liu
- Department of Anatomy, School of Basic Medical Sciences, Mudanjiang Medical College, Mudanjiang, People's Republic of China.,Institute of Neural Tissue Engineering, Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Yan-Ming Pan
- Department of Anatomy, School of Basic Medical Sciences, Mudanjiang Medical College, Mudanjiang, People's Republic of China.,Key Laboratory of Cancer Prevention and Treatment of Heilongjiang Province, Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Yun-Shuang Liu
- Department of Medical Imaging, Hongqi Hospital of Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Jia-Hang Hu
- Department of Medical Imaging, Hongqi Hospital of Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Xiao-Dong Zhang
- Department of Infectious Diseases, Hongqi Hospital of Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Da-Wei Zhang
- Department of Anatomy, School of Basic Medical Sciences, Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Ying Wang
- Department of Anatomy, School of Basic Medical Sciences, Mudanjiang Medical College, Mudanjiang, People's Republic of China.,Institute of Neural Tissue Engineering, Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Yu-Kuan Feng
- Department of Anatomy, School of Basic Medical Sciences, Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Jian-Bo Yu
- Key Laboratory of Cancer Prevention and Treatment of Heilongjiang Province, Mudanjiang Medical College, Mudanjiang, People's Republic of China.,Pathology Diagnosis Center, The First Clinical Medical School of Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Yong-Xia Cheng
- Key Laboratory of Cancer Prevention and Treatment of Heilongjiang Province, Mudanjiang Medical College, Mudanjiang, People's Republic of China.,Pathology Diagnosis Center, The First Clinical Medical School of Mudanjiang Medical College, Mudanjiang, People's Republic of China.,Institute of Stem Cells, Mudanjiang Medical College, Mudanjiang, People's Republic of China
| |
Collapse
|
33
|
Limoli PG, Vingolo EM, Limoli C, Nebbioso M. Stem Cell Surgery and Growth Factors in Retinitis Pigmentosa Patients: Pilot Study after Literature Review. Biomedicines 2019; 7:biomedicines7040094. [PMID: 31801246 PMCID: PMC6966474 DOI: 10.3390/biomedicines7040094] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/23/2019] [Accepted: 11/26/2019] [Indexed: 01/03/2023] Open
Abstract
To evaluate whether grafting of autologous mesenchymal cells, adipose-derived stem cells, and platelet-rich plasma into the supracoroideal space by surgical treatment with the Limoli retinal restoration technique (LRRT) can exert a beneficial effect in retinitis pigmentosa (RP) patients. Twenty-one eyes underwent surgery and were divided based on retinal foveal thickness (FT) ≤ 190 or > 190 µm into group A-FT and group B-FT, respectively. The specific LRRT triad was grafted in a deep scleral pocket above the choroid of each eye. At 6-month follow-up, group B showed a non-significant improvement in residual close-up visus and sensitivity at microperimetry compared to group A. After an in-depth review of molecular biology studies concerning degenerative phenomena underlying the etiopathogenesis of retinitis pigmentosa (RP), it was concluded that further research is needed on tapeto-retinal degenerations, both from a clinical and molecular point of view, to obtain better functional results. In particular, it is necessary to increase the number of patients, extend observation timeframes, and treat subjects in the presence of still trophic retinal tissue to allow adequate biochemical and functional catering.
Collapse
Affiliation(s)
- Paolo Giuseppe Limoli
- Low Vision Research Centre of Milan, p.zza Sempione 3, 20145 Milan, Italy; (P.G.L.); (C.L.)
| | - Enzo Maria Vingolo
- Department of Sense Organs, Faculty of Medicine and Odontology, Sapienza University of Rome, p.le A. Moro 5, 00185 Rome, Italy;
| | - Celeste Limoli
- Low Vision Research Centre of Milan, p.zza Sempione 3, 20145 Milan, Italy; (P.G.L.); (C.L.)
| | - Marcella Nebbioso
- Department of Sense Organs, Faculty of Medicine and Odontology, Sapienza University of Rome, p.le A. Moro 5, 00185 Rome, Italy;
- Correspondence: ; Tel.: +39-06-49975422; Fax: +39-06-49975425
| |
Collapse
|
34
|
Abstract
Autologous fat transfer (AFT) is a well-established and safe surgical technique in which autologous fat tissue is injected at a defective site directly after its harvesting and centrifugation. Initially used as an excellent filler for facial enhancement and rejuvenation, AFT has evolved over the years into more complex reconstructive procedures. Autologous fat transfer is now additionally used for correcting and restoring cranial and maxillofacial volume defects resulting from traumas, tumors, or congenital disorders. Apart from being a natural filler, human adipose tissue represents a rich source of mesenchymal stem cells, called adipose-derived stem cells, that exhibit multilineage differentiation potential and secrete several angiogenic and antiapoptotic factors.In this work, the authors will discuss different aspects influencing the final outcome of AFT, on the basis of the major clinical outcomes obtained using this surgical procedure. Starting from preoperative evaluation and planning, donor-recipient sites, and type of anesthesia and infiltration, the discussion will continue by analyzing the methods of adipose tissue harvesting, purification, and processing. Finally, an overview of the type of placement, clinical application, postoperative care, possible complications, fat graft take, longevity, volume maintenance, and future perspectives will be provided.
Collapse
|
35
|
Namgoong S, Lee H, Lee JS, Jeong SH, Han SK, Dhong ES. Comparative Biological Effects of Human Amnion and Chorion Membrane Extracts on Human Adipose-Derived Stromal Cells. J Craniofac Surg 2019; 30:947-954. [PMID: 30817541 DOI: 10.1097/scs.0000000000005393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Although therapies with human amnion/chorion are used to ameliorate acute and chronic wounds, it is unclear which component of the amnion/chorion tissue promotes wound healing. To characterize the comparative effects of amnion and chorion in wound healing, we used human adipose-derived stromal cells to assess cell viability, migration, and gel contraction after treatment with amnion membrane extract (AME) or chorion membrane extract (CME). We then correlated the possible effectors via AME and CME protein profiling, and compared them by enzyme-linked immunosorbent assay (ELISA), western blotting, and immunocytochemistry. Cell viability was significantly increased with 50 and 100 μg/mL AME treatment, but with CME treatment, a significant increase was only observed with 100 μg/mL. With CME treatment, cell migration was 2.22-fold greater than the control, and collagen gels showed 20% greater contraction. Compared to control, the expression levels of α-smooth muscle actin (SMA) and smooth muscle protein 22-alpha (SM22α) increased both with AME and CME treatments, whereas calponin expression decreased. Protein profiling revealed significantly higher tissue inhibitor of metalloproteinase-1 (TIMP-1), interleukin-8, exotoxin, and adiponectin levels in CME than in AME, and ELISA revealed 8-fold higher adiponectin levels in cells treated with CME than those treated with AME. Immunocytochemistry revealed that α-SMA, SM22α, and calponin were significantly higher in CME- than AME-treated cells; however, adiponectin treatment did not enhance α-SMA, SM22α, or calponin expression. In conclusion, amnion and chorion membrane extracts exerted differential effects on proliferation and contraction of human adipose-derived stromal cells. Amnion extract was superior at inducing cell proliferation and migration, whereas CME was superior at inducing cell contraction.
Collapse
Affiliation(s)
- Sik Namgoong
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
36
|
Panta W, Imsoonthornruksa S, Yoisungnern T, Suksaweang S, Ketudat-Cairns M, Parnpai R. Enhanced Hepatogenic Differentiation of Human Wharton's Jelly-Derived Mesenchymal Stem Cells by Using Three-Step Protocol. Int J Mol Sci 2019; 20:ijms20123016. [PMID: 31226809 PMCID: PMC6627410 DOI: 10.3390/ijms20123016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/11/2019] [Accepted: 06/17/2019] [Indexed: 02/06/2023] Open
Abstract
Currently, human Wharton’s jelly-derived mesenchymal stem cells (hWJ-MSCs) are an attractive source of stem cells for cell-based therapy, owing to their ability to undergo self-renewal and differentiate into all mesodermal, some neuroectodermal, and endodermal progenies, including hepatocytes. Herein, this study aimed to investigate the effects of sodium butyrate (NaBu), an epigenetic regulator that directly inhibits histone deacetylase, on hepatic endodermal lineage differentiation of hWJ-MSCs. NaBu, at 1 mM, optimally promoted endodermal differentiation of hWJ-MSCs, along with epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) supplementation. CXCR4, HNF3β, SOX17 (endodermal), and GATA6 (mesendodermal) mRNAs were also up-regulated (p < 0.001). Immunocytochemistry and a Western blot analysis of SOX17 and HNF3β confirmed that the 1 mM NaBu along with EGF and bFGF supplementation condition was appropriately pre-treated with hWJ-MSCs before hepatogenic differentiation. Furthermore, the hepatic differentiation medium with NaBu pre-treatment up-regulated hepatoblast (AFP and HNF3β) and hepatic (CK18 and ALB) markers, and increased the proportion of mature hepatocyte functions, including G6P, C/EBPα, and CYP2B6 mRNAs, glycogen storage and urea secretion. The hepatic differentiation medium with NaBu in the pre-treatment step can induce hWJ-MSC differentiation toward endodermal, hepatoblastic, and hepatic lineages. Therefore, the hepatic differentiation medium with NaBu pre-treatment for differentiating hWJ-MSCs could represent an alternative protocol for cell-based therapy and drug screening in clinical applications.
Collapse
Affiliation(s)
- Wachira Panta
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| | - Sumeth Imsoonthornruksa
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| | - Ton Yoisungnern
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| | - Sanong Suksaweang
- School of Pathology and Laboratory Medicine, Institute of Medicine, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| | - Mariena Ketudat-Cairns
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| | - Rangsun Parnpai
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| |
Collapse
|
37
|
Toosi S, Esmaeilzadeh Z, Naderi‐Meshkin H, Heirani‐Tabasi A, Peivandi MT, Behravan J. Adipocyte lineage differentiation potential of MSCs isolated from reaming material. J Cell Physiol 2019; 234:20066-20071. [DOI: 10.1002/jcp.28605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 03/19/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Shirin Toosi
- Biotechnology Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
| | - Zohreh Esmaeilzadeh
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education Culture and Research (ACECR), Khorasan Razavi Branch Mashhad Iran
| | - Hojjat Naderi‐Meshkin
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education Culture and Research (ACECR), Khorasan Razavi Branch Mashhad Iran
| | - Asieh Heirani‐Tabasi
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education Culture and Research (ACECR), Khorasan Razavi Branch Mashhad Iran
| | - Mohammad Taghi Peivandi
- Department of Orthopedic Surgery, Orthopedic and Trauma Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Javad Behravan
- Biotechnology Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
- School of Pharmacy University of Waterloo Waterloo Ontario Canada
- Center for Bioengineering and Biotechnology University of Waterloo Waterloo Ontario Canada
| |
Collapse
|
38
|
Guo H, Li B, Wang W, Zhao N, Gao H. Mesenchymal stem cells overexpressing IL-35: a novel immunosuppressive strategy and therapeutic target for inducing transplant tolerance. Stem Cell Res Ther 2018; 9:254. [PMID: 30257721 PMCID: PMC6158805 DOI: 10.1186/s13287-018-0988-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Inducing donor-specific immunological tolerance, which avoids the complications of long-term immunosuppression, is an important goal in organ transplantation. Interleukin-35 (IL-35), a cytokine identified in 2007, is mainly secreted by regulatory T cells (Tregs) and is essential for Tregs to exert their maximal immunoregulatory activity in vitro and in vivo. A growing number of studies show that IL-35 plays an important role in autoimmune diseases and infectious diseases. Recent research has shown that IL-35 could effectively alleviate allograft rejection and has the potential to be a novel therapeutic strategy for graft rejection. With increasing study of immunoregulation, cell-based therapy has become a novel approach to attenuate rejection after transplantation. Mesenchymal stem cells (MSCs), which exhibit important properties of multilineage differentiation, tissue repair, and immunoregulation, have recently emerged as attractive candidates for cell-based therapeutics, especially in transplantation. Accumulating evidence demonstrates that the therapeutic abilities of MSCs can be amplified by gene modification. Therefore, researchers have constructed IL-35 gene-modified MSCs and explored their functions and mechanisms in some disease models. In this review, we discuss the potential tolerance-inducing effects of MSCs in transplantation and briefly introduce the immunoregulatory functions of the IL-35 gene-modified MSCs.
Collapse
Affiliation(s)
- Hao Guo
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Baozhu Li
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Wei Wang
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Na Zhao
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Haopeng Gao
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| |
Collapse
|
39
|
Luo L, Hu DH, Yin JQ, Xu RX. Molecular Mechanisms of Transdifferentiation of Adipose-Derived Stem Cells into Neural Cells: Current Status and Perspectives. Stem Cells Int 2018; 2018:5630802. [PMID: 30302094 PMCID: PMC6158979 DOI: 10.1155/2018/5630802] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/12/2018] [Accepted: 07/19/2018] [Indexed: 12/19/2022] Open
Abstract
Neurological diseases can severely compromise both physical and psychological health. Recently, adult mesenchymal stem cell- (MSC-) based cell transplantation has become a potential therapeutic strategy. However, most studies related to the transdifferentiation of MSCs into neural cells have had disappointing outcomes. Better understanding of the mechanisms underlying MSC transdifferentiation is necessary to make adult stem cells more applicable to treating neurological diseases. Several studies have focused on adipose-derived stromal/stem cell (ADSC) transdifferentiation. The purpose of this review is to outline the molecular characterization of ADSCs, to describe the methods for inducing ADSC transdifferentiation, and to examine factors influencing transdifferentiation, including transcription factors, epigenetics, and signaling pathways. Exploring and understanding the mechanisms are a precondition for developing and applying novel cell therapies.
Collapse
Affiliation(s)
- Liang Luo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi 710032, China
- Stem Cell Research Center, Neurosurgery Institute of PLA Army, Beijing 100700, China
- Bayi Brain Hospital, General Hospital of PLA Army, Beijing 100700, China
| | - Da-Hai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shanxi 710032, China
| | - James Q. Yin
- Stem Cell Research Center, Neurosurgery Institute of PLA Army, Beijing 100700, China
- Bayi Brain Hospital, General Hospital of PLA Army, Beijing 100700, China
| | - Ru-Xiang Xu
- Stem Cell Research Center, Neurosurgery Institute of PLA Army, Beijing 100700, China
- Bayi Brain Hospital, General Hospital of PLA Army, Beijing 100700, China
| |
Collapse
|
40
|
Willis GR, Fernandez-Gonzalez A, Reis M, Mitsialis SA, Kourembanas S. Macrophage Immunomodulation: The Gatekeeper for Mesenchymal Stem Cell Derived-Exosomes in Pulmonary Arterial Hypertension? Int J Mol Sci 2018; 19:ijms19092534. [PMID: 30150544 PMCID: PMC6164282 DOI: 10.3390/ijms19092534] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 12/13/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by remodeling of the pulmonary arteries, increased pulmonary infiltrates, loss of vascular cross-sectional area, and elevated pulmonary vascular resistance. Despite recent advances in the management of PAH, there is a pressing need for the development of new tools to effectively treat and reduce the risk of further complications. Dysregulated immunity underlies the development of PAH, and macrophages orchestrate both the initiation and resolution of pulmonary inflammation, thus, manipulation of lung macrophage function represents an attractive target for emerging immunomodulatory therapies, including cell-based approaches. Indeed, mesenchymal stem cell (MSC)-based therapies have shown promise, effectively modulating the macrophage fulcrum to favor an anti-inflammatory, pro-resolving phenotype, which is associated with both histological and functional benefits in preclinical models of pulmonary hypertension (PH). The complex interplay between immune system homeostasis and MSCs remains incompletely understood. Here, we highlight the importance of macrophage function in models of PH and summarize the development of MSC-based therapies, focusing on the significance of MSC exosomes (MEx) and the immunomodulatory and homeostatic mechanisms by which such therapies may afford their beneficial effects.
Collapse
Affiliation(s)
- Gareth R Willis
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Monica Reis
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - S Alex Mitsialis
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Stella Kourembanas
- Division of Newborn Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
41
|
Determining the parameters governing the electrochemical stability of thiols and disulfides self-assembled monolayer on gold electrodes in physiological medium. J Electroanal Chem (Lausanne) 2018. [DOI: 10.1016/j.jelechem.2017.07.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
42
|
Lo Furno D, Mannino G, Giuffrida R, Gili E, Vancheri C, Tarico MS, Perrotta RE, Pellitteri R. Neural differentiation of human adipose-derived mesenchymal stem cells induced by glial cell conditioned media. J Cell Physiol 2018; 233:7091-7100. [PMID: 29737535 DOI: 10.1002/jcp.26632] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/30/2018] [Indexed: 12/15/2022]
Abstract
Adipose-derived mesenchymal stem cells (ASCs) may transdifferentiate into cells belonging to mesodermal, endodermal, and ectodermal lineages. The aim of this study was to verify whether a neural differentiation of ASCs could be induced by a conditioned medium (CM) obtained from cultures of olfactory ensheathing cells (OECs) or Schwann cells (SCs). ASCs were isolated from the stromal vascular fraction of adipose tissue and expanded for 2-3 passages. They were then cultured in OEC-CM or SC-CM for 24 hr or 7 days. At each stage, the cells were tested by immunocytochemistry and flow cytometer analysis to evaluate the expression of typical neural markers such as Nestin, PGP 9.5, MAP2, Synapsin I, and GFAP. Results show that both conditioned media induced similar positive effects, as all tested markers were overexpressed, especially at day 7. Overall, an evident trend toward neuronal or glial differentiation was not clearly detectable in many cases. Nevertheless, a higher tendency toward a neuronal phenotype was recognized for OEC-CM (considering MAP2 increases). On the other hand, SC-CM would be responsible for a more marked glial induction (considering GFAP increases). These findings confirm that environmental features can induce ASCs toward a neural differentiation, either as neuronal or glial elements. Rather than supplementing the culture medium by adding chemical agents, a "more physiological" condition was obtained here by means of soluble factors (cytokines/growth factors) likely released by glial cells. This culture strategy may provide valuable information in the development of cell-based therapeutic approaches for pathologies affecting the central/peripheral nervous system.
Collapse
Affiliation(s)
- Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Giuliana Mannino
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Elisa Gili
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Carlo Vancheri
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Maria S Tarico
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Rosario E Perrotta
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Rosalia Pellitteri
- Institute of Neurological Sciences, National Research Council, Section of Catania, Catania, Italy
| |
Collapse
|
43
|
Abstract
With the advancements in antenatal steroid therapies and surfactant replacement, current clinical practices in neonatal intensive care units allow the survival of infants at very low gestational age. Despite these advances, there continues to be significant morbidity associated with extreme preterm birth that includes both short-term and long-term cardiorespiratory impairment. With no effective single therapy in preventing or treating developmental lung injuries, the need for new tools to treat and reduce risk of complications associated with extreme preterm birth is urgent. Stem cell-based therapies, in particular therapies utilizing mesenchymal stem (stromal) cells (MSCs), have shown promise in a number of animal models of lung pathologies relevant to neonatology. Recent studies in this field have consolidated the concept that the therapeutic mechanism of MSC action is paracrine, and this led to wide acceptance of the concept that the delivery of the MSC secretome rather than live cells may provide an alternative therapeutic approach for many complex diseases. Here, we summarize the significance and application of cell-free based therapies in preclinical models of neonatal lung injury. We emphasize the development of extracellular vesicle (EV)-based therapeutics and focus on the challenges that remain to be addressed before their application to clinical practice.
Collapse
|
44
|
Mesenchymal Stem Cells Form 3D Clusters Following Intraventricular Transplantation. J Mol Neurosci 2018; 65:60-73. [PMID: 29705933 DOI: 10.1007/s12031-018-1070-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/19/2018] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) are regarded as an immune privileged cell type with numerous regeneration-promoting effects. The in vivo behavior of MSC and underlying mechanisms leading to their regenerative effects are largely unknown. The aims of this study were to comparatively investigate the in vivo behavior of canine (cMSC), human (hMSC), and murine MSC (mMSC) following intra-cerebroventricular transplantation. At 7 days post transplantation (dpt), clusters of cMSC, hMSC, and mMSC were detected within the ventricular system. At 49 dpt, cMSC-transplanted mice showed clusters mostly consisting of extracellular matrix lacking transplanted MSC. Similarly, hMSC-transplanted mice lacked MSC clusters at 49 dpt. Xenogeneic MSC transplantation was associated with a local T lymphocyte-dominated immune reaction at both time points. Interestingly, no associated inflammation was observed following syngeneic mMSC transplantation. In conclusion, transplanted MSC formed intraventricular cell clusters and exhibited a short life span in vivo. Xenogeneically in contrast to syngeneically transplanted MSC triggered a T cell-mediated graft rejection indicating that MSCs are not as immune privileged as previously assumed. However, MSC may mediate their effects by a "hit and run" mechanism and future studies will show whether syngeneically or xenogeneically transplanted MSCs exert better therapeutic effects in animals with CNS disease.
Collapse
|
45
|
Song Y, Du H, Dai C, Zhang L, Li S, Hunter DJ, Lu L, Bao C. Human adipose-derived mesenchymal stem cells for osteoarthritis: a pilot study with long-term follow-up and repeated injections. Regen Med 2018; 13:295-307. [PMID: 29417902 DOI: 10.2217/rme-2017-0152] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIM This study aimed to evaluate the safety and therapeutic potential of autologous human adipose-derived mesenchymal stem cells (haMSCs) in patients with osteoarthritis. MATERIALS & METHODS Safety and efficacy of haMSCs were preclinically assessed in vitro and in BALB/c-nu nude mice. 18 patients were enrolled and divided into three dose groups: the low-dose, mid-dose and high-dose group (1 × 107, 2 × 107 and 5 × 107 cells, respectively), provided three injections and followed up for 96 weeks. RESULTS & CONCLUSION The preclinical study established the safety and efficacy of haMSCs. Intra-articular injections of haMSCs were safe and improved pain, function and cartilage volume of the knee joint, rendering them a promising novel treatment for knee osteoarthritis. The dosage of 5 × 107 haMSCs exhibited the highest improvement (ClinicalTrials.gov Identifier: NCT01809769).
Collapse
Affiliation(s)
- Yang Song
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Du
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | - Li Zhang
- Cellular Biomedicine Group Ltd, Shanghai, China
| | - Suke Li
- Cellular Biomedicine Group Ltd, Shanghai, China
| | - David J Hunter
- Rheumatology Department, Royal North Shore Hospital & Institute of Bone & Joint Research, Kolling Institute, University of Sydney, Sydney, Australia
| | - Liangjing Lu
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chunde Bao
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
46
|
"Good things come in small packages": application of exosome-based therapeutics in neonatal lung injury. Pediatr Res 2018; 83:298-307. [PMID: 28985201 PMCID: PMC5876073 DOI: 10.1038/pr.2017.256] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/29/2017] [Indexed: 02/07/2023]
Abstract
Infants born at very low gestational age contribute disproportionately to neonatal morbidity and mortality. Advancements in antenatal steroid therapies and surfactant replacement have favored the survival of infants with ever-more immature lungs. Despite such advances in medical care, cardiopulmonary and neurological impairment prevail in constituting the major adverse outcomes for neonatal intensive care unit survivors. With no single effective therapy for either the prevention or treatment of such neonatal disorders, the need for new tools to treat and reduce risk of further complications associated with extreme preterm birth is urgent. Mesenchymal stem/stromal cell (MSC)-based approaches have shown promise in numerous experimental models of lung injury relevant to neonatology. Recent studies have highlighted that the therapeutic potential of MSCs is harnessed in their secretome, and that the therapeutic vector therein is represented by the exosomes released by MSCs. In this review, we summarize the development and significance of stem cell-based therapies for neonatal diseases, focusing on preclinical models of neonatal lung injury. We emphasize the development of MSC exosome-based therapeutics and comment on the challenges in bringing these promising interventions to clinic.
Collapse
|
47
|
HDAC6 deficiency induces apoptosis in mesenchymal stem cells through p53 K120 acetylation. Biochem Biophys Res Commun 2017; 494:51-56. [DOI: 10.1016/j.bbrc.2017.10.087] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 10/16/2017] [Indexed: 01/05/2023]
|
48
|
Impact of Antibiotics on the Proliferation and Differentiation of Human Adipose-Derived Mesenchymal Stem Cells. Int J Mol Sci 2017; 18:ijms18122522. [PMID: 29186789 PMCID: PMC5751125 DOI: 10.3390/ijms18122522] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue is a promising source of mesenchymal stem cells. Their potential to differentiate and regenerate other types of tissues may be affected by several factors. This may be due to in vitro cell-culture conditions, especially the supplementation with antibiotics. The aim of our study was to evaluate the effects of a penicillin-streptomycin mixture (PS), amphotericin B (AmB), a complex of AmB with copper (II) ions (AmB-Cu2+) and various combinations of these antibiotics on the proliferation and differentiation of adipose-derived stem cells in vitro. Normal human adipose-derived stem cells (ADSC, Lonza) were routinely maintained in a Dulbecco’s Modified Eagle Medium (DMEM) that was either supplemented with selected antibiotics or without antibiotics. The ADSC that were used for the experiment were at the second passage. The effect of antibiotics on proliferation was analyzed using the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) and sulforhodamine-B (SRB) tests. Differentiation was evaluated based on Alizarin Red staining, Oil Red O staining and determination of the expression of ADSC, osteoblast and adipocyte markers by real-time RT-qPCR. The obtained results indicate that the influence of antibiotics on adipose-derived stem cells depends on the duration of exposure and on the combination of applied compounds. We show that antibiotics alter the proliferation of cells and also promote natural osteogenesis, and adipogenesis, and that this effect is also noticeable in stimulated osteogenesis.
Collapse
|
49
|
Willis GR, Kourembanas S, Mitsialis SA. Toward Exosome-Based Therapeutics: Isolation, Heterogeneity, and Fit-for-Purpose Potency. Front Cardiovasc Med 2017; 4:63. [PMID: 29062835 PMCID: PMC5640880 DOI: 10.3389/fcvm.2017.00063] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/25/2017] [Indexed: 12/11/2022] Open
Abstract
Exosomes are defined as submicron (30-150 nm), lipid bilayer-enclosed extracellular vesicles (EVs), specifically generated by the late endosomal compartment through fusion of multivesicular bodies with the plasma membrane. Produced by almost all cells, exosomes were originally considered to represent just a mechanism for jettisoning unwanted cellular moieties. Although this may be a major function in most cells, evolution has recruited the endosomal membrane-sorting pathway to duties beyond mere garbage disposal, one of the most notable examples being its cooption by retroviruses for the generation of Trojan virions. It is, therefore, tempting to speculate that certain cell types have evolved an exosome subclass active in intracellular communication. We term this EV subclass "signalosomes" and define them as exosomes that are produced by the "signaling" cells upon specific physiological or environmental cues and harbor cargo capable of modulating the programming of recipient cells. Our recent studies have established that signalosomes released by mesenchymal stem/stromal cells (MSCs) represent the main vector of MSC immunomodulation and therapeutic action in animal models of lung disease. The efficacy of MSC-exosome treatments in a number of preclinical models of cardiovascular and pulmonary disease supports the promise of application of exosome-based therapeutics across a wide range of pathologies within the near future. However, the full realization of exosome therapeutic potential has been hampered by the absence of standardization in EV isolation, and procedures for purification of signalosomes from the main exosome population. This is mainly due to immature methodologies for exosome isolation and characterization and our incomplete understanding of the specific characteristics and molecular composition of signalosomes. In addition, difficulties in defining metrics for potency of exosome preparations and the challenges of industrial scale-up and good manufacturing practice compliance have complicated smooth and timely transition to clinical development. In this manuscript, we focus on cell culture conditions, exosome harvesting, dosage, and exosome potency, providing some empirical guidance and perspectives on the challenges in bringing exosome-based therapies to clinic.
Collapse
Affiliation(s)
- Gareth R Willis
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Stella Kourembanas
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - S Alex Mitsialis
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
50
|
Abbasalizadeh S, Pakzad M, Cabral JMS, Baharvand H. Allogeneic cell therapy manufacturing: process development technologies and facility design options. Expert Opin Biol Ther 2017; 17:1201-1219. [PMID: 28699788 DOI: 10.1080/14712598.2017.1354982] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Currently, promising outcomes from clinical trials of allogeneic cells, especially allogeneic mesenchymal stromal cells, fibroblasts, keratinocytes, and human cardiac stem cells, have encouraged research institutions, small and medium enterprises (SMEs), and big pharmaceutical companies to invest and focus on developing allogeneic cell therapy products. Commercial and large-scale production of allogeneic cell therapy products requires unique capabilities to develop technologies that generate safe and effective allogeneic cells/cell lines and their fully characterized master/working banks. In addition, it is necessary to design robust upstream and downstream manufacturing processes, and establish integrated, well-designed manufacturing facilities to produce high quality affordable products in accordance with current GMP regulations for the production of cell therapy products. Areas covered: The authors highlight: the recent advances in the development of allogeneic products, the available options to develop robust manufacturing processes, and facility design considerations. Expert opinion: Currently, there are multiple challenges in development of allogeneic cell therapy products. Indeed, the field is still in its infancy; with technologies and regulations still under development, as is our understanding of the mechanisms of action in the body and their interaction with the host immune system. Their characterization and testing is also an emerging and very complex area.
Collapse
Affiliation(s)
- Saeed Abbasalizadeh
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
- b Department of Bioengineering and Institute for Bioengineering and Biosciences , Instituto Superior Técnico, Universidade de Lisboa , Lisboa , Portugal
| | - Mohammad Pakzad
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
| | - Joaquim M S Cabral
- b Department of Bioengineering and Institute for Bioengineering and Biosciences , Instituto Superior Técnico, Universidade de Lisboa , Lisboa , Portugal
| | - Hossein Baharvand
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
- c Department of Developmental Biology , University of Science and Culture , Tehran , Iran
| |
Collapse
|