51
|
Zhou L, Kodidela S, Godse S, Thomas-Gooch S, Kumar A, Raji B, Zhi K, Kochat H, Kumar S. Targeted Drug Delivery to the Central Nervous System Using Extracellular Vesicles. Pharmaceuticals (Basel) 2022; 15:358. [PMID: 35337155 PMCID: PMC8950604 DOI: 10.3390/ph15030358] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/07/2022] [Accepted: 03/07/2022] [Indexed: 02/06/2023] Open
Abstract
The blood brain barrier (BBB) maintains the homeostasis of the central nervous system (CNS) and protects the brain from toxic substances present in the circulating blood. However, the impermeability of the BBB to drugs is a hurdle for CNS drug development, which hinders the distribution of the most therapeutic molecules into the brain. Therefore, scientists have been striving to develop safe and effective technologies to advance drug penetration into the CNS with higher targeting properties and lower off-targeting side effects. This review will discuss the limitation of artificial nanomedicine in CNS drug delivery and the use of natural extracellular vesicles (EVs), as therapeutic vehicles to achieve targeted delivery to the CNS. Information on clinical trials regarding CNS targeted drug delivery using EVs is very limited. Thus, this review will also briefly highlight the recent clinical studies on targeted drug delivery in the peripheral nervous system to shed light on potential strategies for CNS drug delivery. Different technologies engaged in pre- and post-isolation have been implemented to further utilize and optimize the natural property of EVs. EVs from various sources have also been applied in the engineering of EVs for CNS targeted drug delivery in vitro and in vivo. Here, the future feasibility of those studies in clinic will be discussed.
Collapse
Affiliation(s)
- Lina Zhou
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave, Memphis, TN 38163, USA; (L.Z.); (S.G.); (S.T.-G.); (A.K.)
| | - Sunitha Kodidela
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave, Memphis, TN 38163, USA; (L.Z.); (S.G.); (S.T.-G.); (A.K.)
| | - Sandip Godse
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave, Memphis, TN 38163, USA; (L.Z.); (S.G.); (S.T.-G.); (A.K.)
| | - Stacey Thomas-Gooch
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave, Memphis, TN 38163, USA; (L.Z.); (S.G.); (S.T.-G.); (A.K.)
| | - Asit Kumar
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave, Memphis, TN 38163, USA; (L.Z.); (S.G.); (S.T.-G.); (A.K.)
| | - Babatunde Raji
- Plough Center for Sterile Drug Delivery Solutions, University of Tennessee Health Science Center, 208 South Dudley Street, Memphis, TN 38163, USA; (B.R.); (K.Z.); (H.K.)
| | - Kaining Zhi
- Plough Center for Sterile Drug Delivery Solutions, University of Tennessee Health Science Center, 208 South Dudley Street, Memphis, TN 38163, USA; (B.R.); (K.Z.); (H.K.)
| | - Harry Kochat
- Plough Center for Sterile Drug Delivery Solutions, University of Tennessee Health Science Center, 208 South Dudley Street, Memphis, TN 38163, USA; (B.R.); (K.Z.); (H.K.)
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave, Memphis, TN 38163, USA; (L.Z.); (S.G.); (S.T.-G.); (A.K.)
| |
Collapse
|
52
|
Pieters BCH, Arntz OJ, Aarts J, Feitsma AL, van Neerven RJJ, van der Kraan PM, Oliveira MC, van de Loo FAJ. Bovine Milk-Derived Extracellular Vesicles Inhibit Catabolic and Inflammatory Processes in Cartilage from Osteoarthritis Patients. Mol Nutr Food Res 2022; 66:e2100764. [PMID: 34965027 PMCID: PMC9285407 DOI: 10.1002/mnfr.202100764] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 12/17/2021] [Indexed: 11/05/2022]
Abstract
SCOPE Data from the Osteoarthritis Initiative shows that females who drink milk regularly have less joint cartilage loss and OA progression, but the biologic mechanism is unclear. Bovine milk is a rich source of extracellular vesicles (EVs), which are small phospholipid bilayer bound structures that facilitate intercellular communication. In this study, the authors aim to evaluate whether these EVs may have the capacity to protect cartilage from osteoarthritis patients, ex vivo, by directly effecting chondrocytes. METHODS AND RESULTS Human cartilage explants are exposed to cow's milk-derived EVs (CMEVs), which results in reduced sulfated glycosaminoglycan release and inhibition of metalloproteinase-1 expression. Incubation of articular chondrocytes with CMEVs also effectively reduces expression of cartilage destructive enzymes (ADAMTS5, MMPs), which play key roles in the disease progression. In part, these findings are attributed to the presence of TGFβ on these vesicles, and in addition, a possible role is reserved for miR-148a, which is functionally transferred by CMEVs. CONCLUSION These findings highlight the therapeutic potential of local CMEV delivery in osteoarthritic joints, where inflammatory and catabolic mediators are responsible for joint pathology. CMEVs are carriers of both TGFβ and miR-148a, two essential regulators for maintaining chondrocyte homeostasis and protection against cartilage destruction.
Collapse
Affiliation(s)
| | - Onno J. Arntz
- Department of RheumatologyRadboud University Medical CenterNijmegenNetherlands
| | - Joyce Aarts
- Department of RheumatologyRadboud University Medical CenterNijmegenNetherlands
| | | | - R. J. Joost van Neerven
- FrieslandCampinaAmersfoortThe Netherlands
- Cell Biology and ImmunologyWageningen University & ResearchWageningenthe Netherlands
| | | | - Marina C. Oliveira
- Department of NutritionNursing SchoolUniversidade Federal de Minas GeraisBelo HorizonteMinas GeraisBrazil
| | | |
Collapse
|
53
|
DiStefano TJ, Vaso K, Danias G, Chionuma HN, Weiser JR, Iatridis JC. Extracellular Vesicles as an Emerging Treatment Option for Intervertebral Disc Degeneration: Therapeutic Potential, Translational Pathways, and Regulatory Considerations. Adv Healthc Mater 2022; 11:e2100596. [PMID: 34297485 PMCID: PMC8783929 DOI: 10.1002/adhm.202100596] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/08/2021] [Indexed: 12/14/2022]
Abstract
Emergent approaches in regenerative medicine look toward the use of extracellular vesicles (EVs) as a next-generation treatment strategy for intervertebral disc (IVD) degeneration (IVDD) because of their ability to attenuate chronic inflammation, reduce apoptosis, and stimulate proliferation in a number of tissue systems. Yet, there are no Food and Drug Administration (FDA)-approved EV therapeutics in the market with an indication for IVDD, which motivates this article to review the current state of the field and provide an IVD-specific framework to assess its efficacy. In this systematic review, 29 preclinical studies that investigate EVs in relation to the IVD are identified, and additionally, the regulatory approval process is reviewed in an effort to accelerate emerging EV-based therapeutics toward FDA submission and timeline-to-market. The majority of studies focus on nucleus pulposus responses to EV treatment, where the main findings show that stem cell-derived EVs can decelerate the progression of IVDD on the molecular, cellular, and organ level. The findings also highlight the importance of the EV parent cell's pathophysiological and differentiation state, which affects downstream treatment responses and therapeutic outcomes. This systematic review substantiates the use of EVs as a promising cell-free strategy to treat IVDD and enhance endogenous repair.
Collapse
Affiliation(s)
- Tyler J. DiStefano
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Keti Vaso
- Department of Chemical Engineering, The Cooper Union for the Advancement of Science and Art, New York NY, USA
| | - George Danias
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Henry N. Chionuma
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Jennifer R. Weiser
- Department of Chemical Engineering, The Cooper Union for the Advancement of Science and Art, New York NY, USA
| | - James C. Iatridis
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York NY, USA
| |
Collapse
|
54
|
Treatment of Chronic Kidney Disease with Extracellular Vesicles from Mesenchymal Stem Cells and CD133 + Expanded Cells: A Comparative Preclinical Analysis. Int J Mol Sci 2022; 23:ijms23052521. [PMID: 35269664 PMCID: PMC8910174 DOI: 10.3390/ijms23052521] [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: 12/20/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 01/06/2023] Open
Abstract
Chronic kidney disease (CKD) is characterized by structural abnormalities and the progressive loss of kidney function. Extracellular vesicles (EVs) from human umbilical cord tissue (hUCT)-derived mesenchymal stem cells (MSCs) and expanded human umbilical cord blood (hUCB)-derived CD133+ cells (eCD133+) maintain the characteristics of the parent cells, providing a new form of cell-free treatment. We evaluated the effects of EVs from hUCT-derived MSCs and hUCB-derived CD133+ cells on rats with CDK induced by an adenine-enriched diet. EVs were isolated by ultracentrifugation and characterized by nanoparticle tracking analysis (NTA) and electron microscopy. The animals were randomized and divided into the MSC-EV group, eEPC-EV group and control group. Infusions occurred on the seventh and 14th days after CKD induction. Evaluations of kidney function were carried out by biochemical and histological analyses. Intense labeling of the α-SMA protein was observed when comparing the control with MSC-EVs. In both groups treated with EVs, a significant increase in serum albumin was observed, and the increase in cystatin C was inhibited. The results indicated improvements in renal function in CKD, demonstrating the therapeutic potential of EVs derived from MSCs and eCD133+ cells and suggesting the possibility that in the future, more than one type of EV will be used concurrently.
Collapse
|
55
|
del Campo CV, Liaw NY, Gunadasa-Rohling M, Matthaei M, Braga L, Kennedy T, Salinas G, Voigt N, Giacca M, Zimmermann WH, Riley PR. Regenerative potential of epicardium-derived extracellular vesicles mediated by conserved miRNA transfer. Cardiovasc Res 2022; 118:597-611. [PMID: 33599250 PMCID: PMC8803084 DOI: 10.1093/cvr/cvab054] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 02/12/2021] [Indexed: 12/19/2022] Open
Abstract
AIMS After a myocardial infarction, the adult human heart lacks sufficient regenerative capacity to restore lost tissue, leading to heart failure progression. Finding novel ways to reprogram adult cardiomyocytes into a regenerative state is a major therapeutic goal. The epicardium, the outermost layer of the heart, contributes cardiovascular cell types to the forming heart and is a source of trophic signals to promote heart muscle growth during embryonic development. The epicardium is also essential for heart regeneration in zebrafish and neonatal mice and can be reactivated after injury in adult hearts to improve outcome. A recently identified mechanism of cell-cell communication and signalling is that mediated by extracellular vesicles (EVs). Here, we aimed to investigate epicardial signalling via EV release in response to cardiac injury and as a means to optimize cardiac repair and regeneration. METHODS AND RESULTS We isolated epicardial EVs from mouse and human sources and targeted the cardiomyocyte population. Epicardial EVs enhanced proliferation in H9C2 cells and in primary neonatal murine cardiomyocytes in vitro and promoted cell cycle re-entry when injected into the injured area of infarcted neonatal hearts. These EVs also enhanced regeneration in cryoinjured engineered human myocardium (EHM) as a novel model of human myocardial injury. Deep RNA-sequencing of epicardial EV cargo revealed conserved microRNAs (miRs) between human and mouse epicardial-derived exosomes, and the effects on cell cycle re-entry were recapitulated by administration of cargo miR-30a, miR-100, miR-27a, and miR-30e to human stem cell-derived cardiomyocytes and cryoinjured EHM constructs. CONCLUSION Here, we describe the first characterization of epicardial EV secretion, which can signal to promote proliferation of cardiomyocytes in infarcted mouse hearts and in a human model of myocardial injury, resulting in enhanced contractile function. Analysis of exosome cargo in mouse and human identified conserved pro-regenerative miRs, which in combination recapitulated the therapeutic effects of promoting cardiomyocyte proliferation.
Collapse
Affiliation(s)
- Cristina Villa del Campo
- Department of Physiology, Anatomy and Genetics, British Heart Foundation, Oxbridge Centre of Regenerative Medicine, University of Oxford, Sherrington Building, Sherrington Rd, Oxford OX1 3PT, UK
| | - Norman Y Liaw
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Robert-Koch-Straße 42a, 37075 Göttingen, Germany
| | - Mala Gunadasa-Rohling
- Department of Physiology, Anatomy and Genetics, British Heart Foundation, Oxbridge Centre of Regenerative Medicine, University of Oxford, Sherrington Building, Sherrington Rd, Oxford OX1 3PT, UK
| | - Moritz Matthaei
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Luca Braga
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Località Padriciano, 99, 34149 Trieste TS, Italy
- School of Cardiovascular Medicine & Sciences, British Heart Foundation Centre, King's College London, Strand, London WC2R 2L, UK
| | - Tahnee Kennedy
- Department of Physiology, Anatomy and Genetics, British Heart Foundation, Oxbridge Centre of Regenerative Medicine, University of Oxford, Sherrington Building, Sherrington Rd, Oxford OX1 3PT, UK
| | - Gabriela Salinas
- NGS- Integrative Genomics Core Unit (NIG), Institute of Human Genetics, University Medical Centre Göttingen (UMG), Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Robert-Koch-Straße 42a, 37075 Göttingen, Germany
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Località Padriciano, 99, 34149 Trieste TS, Italy
- School of Cardiovascular Medicine & Sciences, British Heart Foundation Centre, King's College London, Strand, London WC2R 2L, UK
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Robert-Koch-Straße 42a, 37075 Göttingen, Germany
| | - Paul Richard Riley
- Department of Physiology, Anatomy and Genetics, British Heart Foundation, Oxbridge Centre of Regenerative Medicine, University of Oxford, Sherrington Building, Sherrington Rd, Oxford OX1 3PT, UK
| |
Collapse
|
56
|
Man K, Barroso IA, Brunet MY, Peacock B, Federici AS, Hoey DA, Cox SC. Controlled Release of Epigenetically-Enhanced Extracellular Vesicles from a GelMA/Nanoclay Composite Hydrogel to Promote Bone Repair. Int J Mol Sci 2022; 23:832. [PMID: 35055017 PMCID: PMC8775793 DOI: 10.3390/ijms23020832] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 02/01/2023] Open
Abstract
Extracellular vesicles (EVs) have garnered growing attention as promising acellular tools for bone repair. Although EVs' potential for bone regeneration has been shown, issues associated with their therapeutic potency and short half-life in vivo hinders their clinical utility. Epigenetic reprogramming with the histone deacetylase inhibitor Trichostatin A (TSA) has been reported to promote the osteoinductive potency of osteoblast-derived EVs. Gelatin methacryloyl (GelMA) hydrogels functionalised with the synthetic nanoclay laponite (LAP) have been shown to effectively bind, stabilise, and improve the retention of bioactive factors. This study investigated the potential of utilising a GelMA-LAP hydrogel to improve local retention and control delivery of epigenetically enhanced osteoblast-derived EVs as a novel bone repair strategy. LAP was found to elicit a dose-dependent increase in GelMA compressive modulus and shear-thinning properties. Incorporation of the nanoclay was also found to enhance shape fidelity when 3D printed compared to LAP-free gels. Interestingly, GelMA hydrogels containing LAP displayed increased mineralisation capacity (1.41-fold) (p ≤ 0.01) over 14 days. EV release kinetics from these nanocomposite systems were also strongly influenced by LAP concentration with significantly more vesicles being released from GelMA constructs as detected by a CD63 ELISA (p ≤ 0.001). EVs derived from TSA-treated osteoblasts (TSA-EVs) enhanced proliferation (1.09-fold), migration (1.83-fold), histone acetylation (1.32-fold) and mineralisation (1.87-fold) of human bone marrow stromal cells (hBMSCs) when released from the GelMA-LAP hydrogel compared to the untreated EV gels (p ≤ 0.01). Importantly, the TSA-EV functionalised GelMA-LAP hydrogel significantly promoted encapsulated hBMSCs extracellular matrix collagen production (≥1.3-fold) and mineralisation (≥1.78-fold) in a dose-dependent manner compared to untreated EV constructs (p ≤ 0.001). Taken together, these findings demonstrate the potential of combining epigenetically enhanced osteoblast-derived EVs with a nanocomposite photocurable hydrogel to promote the therapeutic efficacy of acellular vesicle approaches for bone regeneration.
Collapse
Affiliation(s)
- Kenny Man
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK; (K.M.); (I.A.B.); (M.Y.B.)
| | - Inês A. Barroso
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK; (K.M.); (I.A.B.); (M.Y.B.)
| | - Mathieu Y. Brunet
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK; (K.M.); (I.A.B.); (M.Y.B.)
| | | | - Angelica S. Federici
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland; (A.S.F.); (D.A.H.)
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Trinity College Dublin & RCSI, D02 R590 Dublin, Ireland
| | - David A. Hoey
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland; (A.S.F.); (D.A.H.)
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Trinity College Dublin & RCSI, D02 R590 Dublin, Ireland
| | - Sophie C. Cox
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK; (K.M.); (I.A.B.); (M.Y.B.)
| |
Collapse
|
57
|
Xiao Y, Zhang Y, Li Y, Peng N, Liu Q, Qiu D, Cho J, Borlongan CV, Yu G. Exosomes Derived From Mesenchymal Stem Cells Pretreated With Ischemic Rat Heart Extracts Promote Angiogenesis via the Delivery of DMBT1. Cell Transplant 2022; 31:9636897221102898. [PMID: 35726847 PMCID: PMC9218457 DOI: 10.1177/09636897221102898] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/26/2022] [Accepted: 05/09/2022] [Indexed: 12/02/2022] Open
Abstract
Mesenchymal stem cell-derived exosomes (MSC-Exos) have been shown to promote angiogenesis. Treating MSCs with ischemic rat brain extracts was sufficient to augment their benefits in stroke. However, no similar analyses of ischemic heart extracts have been performed to date. We aim to determine whether MSC-Exos derived from MSCs pretreated with ischemic rat heart extract were able to promote angiogenesis and to clarify underlying mechanisms. ELISA (enzyme-linked immunosorbent assay) of heart extracts revealed a significant increase of vascular endothelial growth factor (VEGF) at 24 h post-MI (myocardial infarction) modeling, and time-dependent decreases in hypoxia inducible factor-1α (HIF-1α). MTT and wound healing assays revealed human umbilical vein endothelial cells (HUVECs) migration and proliferation increased following MSCE-Exo treatment (exosomes derived from MSC pretreated with ischemic heart extracts of 24 h post-MI) relative to MSCN-Exo treatment (exosomes derived from MSC pretreated with normal heart extracts). Proteomic analyses of MSCE-Exo and MSCN-Exo were conducted to screen for cargo proteins promoting angiogenesis. Result revealed several angiogenesis-related proteins were upregulated in MSCE-Exo, including DMBT1 (deleted in malignant brain tumors 1). When DMBT1 was silenced in MSCs, HUVECs with MSCDMBT1 RNAi-Exo treatment exhibited impaired proliferative and migratory activity and reductions of DMBT1, p-Akt, β-catenin, and VEGF. To explore how ischemic heart extracts took effects, ELISA was conducted showing a significant increase of IL-22 at 24 h post-MI modeling. P-STAT3, IL22RA1, DMBT1, and VEGF proteins were increased in MSCE relative to MSCN, and VEGF and DMBT1 were increased in MSCE-Exos. Together, these suggest that IL-22 upregulation in ischemic heart extracts can increase DMBT1 in MSCs. Exosomes derived from those MSCs deliver DMBT1 to HUVECs, thereby enhancing their migratory and proliferative activity.
Collapse
Affiliation(s)
- Yi Xiao
- Division of Cardiovascular, Xiangya Hospital, Central South University, Changsha, China
| | - Ye Zhang
- Division of Cardiovascular, Xiangya Hospital, Central South University, Changsha, China
| | - Yuzhang Li
- Division of Cardiovascular, Xiangya Hospital, Central South University, Changsha, China
| | - Nanyin Peng
- Division of Cardiovascular, Xiangya Hospital, Central South University, Changsha, China
| | - Qin Liu
- Division of Cardiovascular, Xiangya Hospital, Central South University, Changsha, China
| | - Danyang Qiu
- Division of Cardiovascular, Xiangya Hospital, Central South University, Changsha, China
| | - Justin Cho
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Cesario V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Guolong Yu
- Division of Cardiovascular, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
58
|
Prospects and Applications of Natural Blood-Derived Products in Regenerative Medicine. Int J Mol Sci 2021; 23:ijms23010472. [PMID: 35008900 PMCID: PMC8745602 DOI: 10.3390/ijms23010472] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/21/2021] [Accepted: 12/28/2021] [Indexed: 12/16/2022] Open
Abstract
Currently, there are a number of therapeutic schemes used for the treatment of various types of musculoskeletal disorders. However, despite the use of new treatment options, therapeutic failure remains common due to impaired and delayed healing, or implant rejection. Faced with this challenge, in recent years regenerative medicine started looking for alternative solutions that could additionally support tissue regeneration. This review aims to outline the functions and possible clinical applications of, and future hopes associated with, using autologous or heterologous products such as antimicrobial peptides (AMPs), microvesicles (MVs), and neutrophil degranulation products (DGP) obtained from circulating neutrophils. Moreover, different interactions between neutrophils and platelets are described. Certain products released from neutrophils are critical for interactions between different immune cells to ensure adequate tissue repair. By acting directly and indirectly on host cells, these neutrophil-derived products can modulate the body’s inflammatory responses in various ways. The development of new formulations based on these products and their clinically proven success would give hope for significant progress in regenerative therapy in human and veterinary medicine.
Collapse
|
59
|
Luiz RDS, Rampaso RR, Dos Santos AAC, Convento MB, Barbosa DA, da Fonseca CD, de Oliveira AS, Caires A, Furlan A, Schor N, Borges FT. BM-MSC-derived small extracellular vesicles (sEV) from trained animals presented nephroprotective potential in unilateralureteral obstruction model. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20200187. [PMID: 34925478 PMCID: PMC8650265 DOI: 10.1590/1678-9199-jvatitd-2020-0187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 05/21/2021] [Indexed: 12/03/2022] Open
Abstract
Background: The efficacy of bone marrow mesenchymal stromal cells (BM-MSC) and its extracellular vesicles has been demonstrated for a broad spectrum of indications, including kidney diseases. However, BM-MSC donor characteristics and their potential are not usually considered. Therefore, the present work aims to evaluate the nephroprotective capacity of sEV secreted by BM-MSC from trained rats inunilateral ureteral obstruction (UUO) model. Methods: BM-MSC was characterized by their differentiation potential and immunophenotypic markers. The sEV were isolated by ultracentrifugation and characterized by nanoparticle tracking analysis and western blot. Its miRNA cargo was examined by quantitative PCR analysis for miR-26a, 126a, and 296. Wistar rats were submitted to UUO procedure and concomitantly treated with sEV secreted by BM-MSC from the untrained andtrained rats. The kidney tissue from all groups was evaluated for fibrosis mediators (transforming growth factor beta1 and collagen), CD34-angiogenesis marker, and hypoxia-inducible factor 1 alpha (HIF-1α). Results: Treadmill training stimulated in BM-MSC the production of sEV loaded with pro-angiogenic miR-296. The treatment with this sEVin UUO-rats was able to attenuate collagen accumulation and increase CD34 and HIF-1α in the kidney tissue when compared to untrained ones. Tubular proximal cells under hypoxia and exposed to BM-MSC sEV demonstrate accumulation in HIF-1α and NFR-2 (nuclear factor erythroid 2-related factor 2), possibly to mediate the response to hypoxia and oxidative stress, under these conditions. Conclusion: The BM-MSC sEV from trained animals presented an increased nephroprotective potential compared to untrained vesicles by carrying 296-angiomiR and contributing to angiogenesis in UUO model.
Collapse
Affiliation(s)
- Rafael da Silva Luiz
- Nephrology Division, Department of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Rodolfo Rosseto Rampaso
- Nephrology Division, Department of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Alef Aragão Carneiro Dos Santos
- Interdisciplinary Program in Health Sciences, Institute of Physical Activity and Sport Sciences, Cruzeiro do Sul University, São Paulo, SP, Brazil
| | - Marcia Bastos Convento
- Nephrology Division, Department of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Dulce Aparecida Barbosa
- Paulista School of Nursing, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | | | - Andréia Silva de Oliveira
- Nephrology Division, Department of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Agnaldo Caires
- Nephrology Division, Department of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Andrei Furlan
- Nephrology Division, Department of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Nestor Schor
- Nephrology Division, Department of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Fernanda Teixeira Borges
- Nephrology Division, Department of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil.,Interdisciplinary Program in Health Sciences, Institute of Physical Activity and Sport Sciences, Cruzeiro do Sul University, São Paulo, SP, Brazil
| |
Collapse
|
60
|
Small Extracellular Vesicles Derived from Human Chorionic MSCs as Modern Perspective towards Cell-Free Therapy. Int J Mol Sci 2021; 22:ijms222413581. [PMID: 34948379 PMCID: PMC8706681 DOI: 10.3390/ijms222413581] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are of great interest to scientists due to their application in cell therapy of many diseases, as well as regenerative medicine and tissue engineering. Recently, there has been growing evidence surrounding the research based on extracellular vesicles (EVs), especially small EVs (sEVs)/exosomes derived from MSCs. EVs/exosomes can be secreted by almost all cell types and various types of EVs show multiple functions. In addition, MSCs-derived exosomes have similar characteristics and biological activities to MSCs and their therapeutic applications are considered as a safe strategy in cell-free therapy. The aim of this study was the characterization of MSCs isolated from the chorion (CHo-MSCs) of human full-term placenta, as well as the isolation and analysis of small EVs obtained from these cells. Accordingly, in this study, the ability of small EVs' uptake is indicated by synovial fibroblasts, osteoblasts and periosteum-derived MSCs. Improvement in the understanding of the structure, characteristics, mechanism of action and potential application of MSCs-derived small EVs can provide new insight into improved therapeutic strategies.
Collapse
|
61
|
Nyam-Erdene A, Nebie O, Delila L, Buée L, Devos D, Chou SY, Blum D, Burnouf T. Characterization and Chromatographic Isolation of Platelet Extracellular Vesicles from Human Platelet Lysates for Applications in Neuroregenerative Medicine. ACS Biomater Sci Eng 2021; 7:5823-5835. [PMID: 34846835 DOI: 10.1021/acsbiomaterials.1c01226] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human platelet lysates (HPLs) made from clinical-grade platelet concentrates are currently evaluated in the preclinical models of Parkinson's disease, Alzheimer's disease, traumatic brain injury, and others, as a new polyvalent neuroprotective biotherapy of the central nervous system. However, the presence and content of extracellular vesicles (EVs) in HPLs and their potential contribution to the neuroprotective and neurorestorative activities of HPLs are still unknown. We, therefore, characterized the EVs present in four different HPL preparations and after purification by size-exclusion chromatography. We then tested the effect of the isolated EVs on neuronal cell repair. We identified that all four HPLs contained a high and similar amount of EVs (1011 to 1012/mL) with a mean size ranging from ca. 50 to 300 nm and a negative zeta potential as determined by nanoparticle tracking analysis and dynamic light scattering. Western blot analysis revealed that the EVs present in HPLs expressed the clusters of differentiation 41 (CD41) and 61 (CD61) characteristic of platelets. These EVs were efficiently isolated from HPL proteins by Sepharose CL-2B size-exclusion column chromatography as confirmed by total protein determination and protein profile by sodium dodecyl sulfate polyacrylamide gel electrophoresis, with 73-85% recovery and maintenance of their size, negative zeta potential, and CD41 and CD61 expression. Interestingly, the EVs purified from the four HPLs exhibited a differential capacity to promote cell growth and migration in a wound-healing assay using SH-SY5Y neuronal cells, and one EV preparation stimulated network formation in primary neuronal cultures. These data indicated that the EVs present in HPLs have different neuroregenerative capacities and that some EV preparations may have interesting applications as a stand-alone therapy for usage in neuroregenerative medicine.
Collapse
Affiliation(s)
- Ariunjargal Nyam-Erdene
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 101, Taiwan
| | - Ouada Nebie
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Liling Delila
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Luc Buée
- Université de Lille, Inserm U1172, CHU-Lille, Lille Neuroscience & Cognition, Lille 59000, France.,Alzheimer & Tauopathies, Labex DISTALZ, Lille 59000, France.,NeuroTMULille International Laboratory, Université de Lille, Lille 59000, France
| | - David Devos
- Université de Lille, Inserm U1172, CHU-Lille, Lille Neuroscience & Cognition, Lille 59000, France.,NeuroTMULille International Laboratory, Université de Lille, Lille 59000, France
| | - Szu-Yi Chou
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.,NeuroTMULille International Laboratory, Taipei Medical University, Taipei 101, Taiwan
| | - David Blum
- Université de Lille, Inserm U1172, CHU-Lille, Lille Neuroscience & Cognition, Lille 59000, France.,Alzheimer & Tauopathies, Labex DISTALZ, Lille 59000, France.,NeuroTMULille International Laboratory, Université de Lille, Lille 59000, France
| | - Thierry Burnouf
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 101, Taiwan.,Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan.,NeuroTMULille International Laboratory, Taipei Medical University, Taipei 101, Taiwan.,International PhD Program in Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.,Brain and Consciousness Research Centre, TMU Shuang Ho Hospital, New Taipei City 106, Taiwan
| |
Collapse
|
62
|
Tejeda-Mora H, Leon LG, Demmers J, Baan CC, Reinders MEJ, Bleck B, Lombardo E, Merino A, Hoogduijn MJ. Proteomic Analysis of Mesenchymal Stromal Cell-Derived Extracellular Vesicles and Reconstructed Membrane Particles. Int J Mol Sci 2021; 22:ijms222312935. [PMID: 34884740 PMCID: PMC8657583 DOI: 10.3390/ijms222312935] [Citation(s) in RCA: 5] [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: 11/12/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EV) derived from mesenchymal stromal cells (MSC) are a potential therapy for immunological and degenerative diseases. However, large-scale production of EV free from contamination by soluble proteins is a major challenge. The generation of particles from isolated membranes of MSC, membrane particles (MP), may be an alternative to EV. In the present study we generated MP from the membranes of lysed MSC after removal of the nuclei. The yield of MP per MSC was 1 × 105 times higher than EV derived from the same number of MSC. To compare the proteome of MP and EV, proteomic analysis of MP and EV was performed. MP contained over 20 times more proteins than EV. The proteins present in MP evidenced a multi-organelle origin of MP. The projected function of the proteins in EV and MP was very different. Whilst proteins in EV mainly play a role in extracellular matrix organization, proteins in MP were interconnected in diverse molecular pathways, including protein synthesis and degradation pathways and demonstrated enzymatic activity. Treatment of MSC with IFNγ led to a profound effect on the protein make up of EV and MP, demonstrating the possibility to modify the phenotype of EV and MP through modification of parent MSC. These results demonstrate that MP are an attractive alternative to EV for the development of potential therapies. Functional studies will have to demonstrate therapeutic efficacy of MP in preclinical disease models.
Collapse
Affiliation(s)
- Hector Tejeda-Mora
- Erasmus MC Transplant Institute, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (H.T.-M.); (C.C.B.); (M.E.J.R.); (A.M.)
| | - Leticia G. Leon
- The Netherlands Cancer Institute, Department of Pathology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands;
| | - Jeroen Demmers
- Proteomics Center, Erasmus MC, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Carla C. Baan
- Erasmus MC Transplant Institute, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (H.T.-M.); (C.C.B.); (M.E.J.R.); (A.M.)
| | - Marlies E. J. Reinders
- Erasmus MC Transplant Institute, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (H.T.-M.); (C.C.B.); (M.E.J.R.); (A.M.)
| | - Bertram Bleck
- Takeda, Gastrointestinal Drug Discovery Unit, Cambridge, MA 02139, USA;
| | - Eleuterio Lombardo
- Takeda Madrid, Cell Therapy Technology Center-Cell Therapies, 28046 Madrid, Spain;
| | - Ana Merino
- Erasmus MC Transplant Institute, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (H.T.-M.); (C.C.B.); (M.E.J.R.); (A.M.)
| | - Martin J. Hoogduijn
- Erasmus MC Transplant Institute, Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (H.T.-M.); (C.C.B.); (M.E.J.R.); (A.M.)
- Correspondence:
| |
Collapse
|
63
|
Liu J, Li F, Liu B, Yao Z, Li L, Liu G, Peng L, Wang Y, Huang J. Adipose-derived mesenchymal stem cell exosomes inhibit transforming growth factor-β1-induced collagen synthesis in oral mucosal fibroblasts. Exp Ther Med 2021; 22:1419. [PMID: 34707701 PMCID: PMC8543178 DOI: 10.3892/etm.2021.10854] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 05/12/2021] [Indexed: 12/20/2022] Open
Abstract
Oral submucosal fibrosis (OSF) is a potentially malignant oral disorder that requires the further development of advanced treatment strategies. TGF-β1 has been reported to be the main trigger for the increased collagen production and reduced activity of matrix degradation pathways in OSF. Exosomes are key mediators of paracrine signaling that have been proposed for direct use as therapeutic agents for tissue repair and regeneration. The present study aimed to investigate the effects of human adipose-derived mesenchymal stem cell (ADSC) exosomes (ADSC-Exos) on TGF-β1-treated oral fibroblasts in vitro and to unravel the potential underlying mechanism of action. Oral mucosal fibroblasts were obtained from the buccal tissues of patients without OSF during extraction of the third molar. ADSCs were obtained from three healthy female individuals during liposuction procedures. ADSC-Exos were isolated by ultracentrifugation and identified by electron microscopy, nanoparticle tracking and western blotting. Immunofluorescence and immunocytochemistry staining were performed to measure the expression levels of vimentin and α-smooth muscle actin in the fibroblasts. Reverse transcription-quantitative PCR and western blotting were used to determine the expression levels of mRNAs and proteins associated with collagen production. The p38 MAPK activator anisomycin was used to identify the underlying mechanisms of the effects of ADSC-Exos on TGF-β1-induced collagen synthesis in oral mucosal fibroblasts. The results of the present study revealed that ADSC-Exos exhibited a cup- or sphere-shaped morphology, with a mean diameter of 58.01±16.17 nm. ADSC-Exos were also found to be positive for CD63 and tumor susceptibility 101 expression. ADSC-Exos treatment reversed the TGF-β1-induced upregulation of collagen I and III protein expression. In addition, in the presence of TGF-β1, the expression levels of collagen type I α 1 chain and collagen type III α 1 chain mRNA were downregulated, whilst the expression levels of matrix metalloproteinase (MMP)1 and MMP3 were upregulated following ADSC-Exos treatment. The TGF-β1-induced upregulation in the phosphorylation of p38 in addition to the increased protein expression of collagens I and III were also reversed in fibroblasts following ADSC-Exos treatment. However, anisomycin treatment alleviated these ADSC-Exos-induced changes. In conclusion, findings from the present study suggest that ADSC-Exos may represent a promising strategy for OSF treatment by targeting the p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Junjie Liu
- Hunan Key Laboratory of Oral Health Research, Hunan Xiangya Stomatological Hospital, Central South University, Changsha, Hunan 410000, P.R. China.,Hunan 3D Printing Engineering Research Center, Hunan Xiangya Stomatological Hospital, Central South University, Changsha, Hunan 410000, P.R. China
| | - Fuxingzi Li
- Department of Endocrinology and Metabolism, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Binjie Liu
- Hunan Key Laboratory of Oral Health Research, Hunan Xiangya Stomatological Hospital, Central South University, Changsha, Hunan 410000, P.R. China
| | - Zhigang Yao
- Hunan Key Laboratory of Oral Health Research, Hunan Xiangya Stomatological Hospital, Central South University, Changsha, Hunan 410000, P.R. China
| | - Long Li
- Hunan Key Laboratory of Oral Health Research, Hunan Xiangya Stomatological Hospital, Central South University, Changsha, Hunan 410000, P.R. China
| | - Gui Liu
- Hunan Key Laboratory of Oral Health Research, Hunan Xiangya Stomatological Hospital, Central South University, Changsha, Hunan 410000, P.R. China.,Hunan 3D Printing Engineering Research Center, Hunan Xiangya Stomatological Hospital, Central South University, Changsha, Hunan 410000, P.R. China
| | - Lei Peng
- Department of Urban Palliative Home Care, Grey Nuns Community Hospital, Edmonton, AB T5J3E4, Canada
| | - Yuxin Wang
- Department of Stomatology, Changsha Central Hospital, Changsha, Hunan 410018, P.R. China
| | - Junhui Huang
- Hunan Key Laboratory of Oral Health Research, Hunan Xiangya Stomatological Hospital, Central South University, Changsha, Hunan 410000, P.R. China.,Hunan 3D Printing Engineering Research Center, Hunan Xiangya Stomatological Hospital, Central South University, Changsha, Hunan 410000, P.R. China
| |
Collapse
|
64
|
Takahashi T, Schleimer RP. Epithelial-Cell-Derived Extracellular Vesicles in Pathophysiology of Epithelial Injury and Repair in Chronic Rhinosinusitis: Connecting Immunology in Research Lab to Biomarkers in Clinics. Int J Mol Sci 2021; 22:11709. [PMID: 34769139 PMCID: PMC8583779 DOI: 10.3390/ijms222111709] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Epithelial barrier disruption and failure of epithelial repair by aberrant epithelial-mesenchymal transition (EMT)-induced basal cells observed in nasal mucosa of chronic rhinosinusitis (CRS) are speculated to play important roles in disease pathophysiology. Microparticles (MPs) are a type of extracellular vesicle (EV) released by budding or shedding from the plasma membrane of activated or apoptotic cells. MPs are detected in nasal lavage fluids (NLFs) and are now receiving attention as potential biomarkers to evaluate the degree of activation of immune cells and injury of structural cells in nasal mucosa of subjects with sinus disease. There are three types of epithelial-cell-derived MPs, which are defined by the expression of different epithelial specific markers on their surface: EpCAM, E-cadherin, and integrin β6 (ITGB6). When these markers are on MPs that are also carrying canonical EMT/mesenchymal markers (Snail (SNAI1); Slug (SNAI2); alpha-smooth muscle actin (αSMA, ACTA2)) or pro- and anti-coagulant molecules (tissue factor (TF); tissue plasminogen activator (tPA); plasminogen activator inhibitor-1 (PAI-1)), they provide insight as to the roles of epithelial activation for EMT or regulation of coagulation in the underlying disease. In this review, we discuss the potential of epithelial MPs as research tools to evaluate status of nasal mucosae of CRS patients in the lab, as well as biomarkers for management and treatment of CRS in the clinic.
Collapse
Affiliation(s)
- Toru Takahashi
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
| | - Robert P Schleimer
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
65
|
Ng CY, Chai JY, Foo JB, Mohamad Yahaya NH, Yang Y, Ng MH, Law JX. Potential of Exosomes as Cell-Free Therapy in Articular Cartilage Regeneration: A Review. Int J Nanomedicine 2021; 16:6749-6781. [PMID: 34621125 PMCID: PMC8491788 DOI: 10.2147/ijn.s327059] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/22/2021] [Indexed: 12/20/2022] Open
Abstract
Treatment of cartilage defects such as osteoarthritis (OA) and osteochondral defect (OCD) remains a huge clinical challenge in orthopedics. OA is one of the most common chronic health conditions and is mainly characterized by the degeneration of articular cartilage, shown in the limited capacity for intrinsic repair. OCD refers to the focal defects affecting cartilage and the underlying bone. The current OA and OCD management modalities focus on symptom control and on improving joint functionality and the patient’s quality of life. Cell-based therapy has been evaluated for managing OA and OCD, and its chondroprotective efficacy is recognized mainly through paracrine action. Hence, there is growing interest in exploiting extracellular vesicles to induce cartilage regeneration. In this review, we explore the in vivo evidence of exosomes on cartilage regeneration. A total of 29 in vivo studies from the PubMed and Scopus databases were identified and analyzed. The studies reported promising results in terms of in vivo exosome delivery and uptake; improved cartilage morphological, histological, and biochemical outcomes; enhanced subchondral bone regeneration; and improved pain behavior following exosome treatment. In addition, exosome therapy is safe, as the included studies documented no significant complications. Modifying exosomal cargos further increased the cartilage and subchondral bone regeneration capacity of exosomes. We conclude that exosome administration is a potent cell-free therapy for alleviating OA and OCD. However, additional studies are needed to confirm the therapeutic potential of exosomes and to identify the standard protocol for exosome-based therapy in OA and OCD management.
Collapse
Affiliation(s)
- Chiew Yong Ng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, 56000, Malaysia
| | - Jia Ying Chai
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, 56000, Malaysia
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, 47500, Selangor, Malaysia.,Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, 47500, Malaysia
| | - Nor Hamdan Mohamad Yahaya
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, 56000, Malaysia
| | - Ying Yang
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Min Hwei Ng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, 56000, Malaysia
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, 56000, Malaysia
| |
Collapse
|
66
|
Grangier A, Branchu J, Volatron J, Piffoux M, Gazeau F, Wilhelm C, Silva AKA. Technological advances towards extracellular vesicles mass production. Adv Drug Deliv Rev 2021; 176:113843. [PMID: 34147532 DOI: 10.1016/j.addr.2021.113843] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/29/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs) are becoming essential actors in bio-therapeutics, as much for their regenerative or immunomodulatory properties as for their potential as cargo delivery vehicles. To enable the democratization of these EV-based therapies, many challenges remain such as large-scale production which is necessary to reduce costs of treatment. Herein, we review some advanced works on high-yield EV manufacturing. One approach consists in developing large-scale cell culture platforms, while others focus on cell stimulation to increase particle yield per cell. This can be done by moderate physico-chemical stresses or by disrupting cell membrane towards autoassembled vesicle-like particles. We critically compare these different techniques, keeping in mind that the field still lacks shared characterization standards, underline the importance of therapeutic potency assessment and discuss mass production strategies that have been identified in current clinical trials.
Collapse
Affiliation(s)
- Alice Grangier
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 75013 and 75006 Paris, France
| | | | | | - Max Piffoux
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 75013 and 75006 Paris, France; Everzom, 75006 Paris, France; Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Florence Gazeau
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 75013 and 75006 Paris, France
| | - Claire Wilhelm
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 75013 and 75006 Paris, France; Laboratoire PhysicoChimie Curie, Institut Curie, PSL Research University - Sorbonne Université - CNRS, 75005 Paris, France.
| | - Amanda K A Silva
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 75013 and 75006 Paris, France.
| |
Collapse
|
67
|
Li Z, Huang Z, Bai L. Cell Interplay in Osteoarthritis. Front Cell Dev Biol 2021; 9:720477. [PMID: 34414194 PMCID: PMC8369508 DOI: 10.3389/fcell.2021.720477] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/14/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a common chronic disease and a significant health concern that needs to be urgently solved. OA affects the cartilage and entire joint tissues, including the subchondral bone, synovium, and infrapatellar fat pads. The physiological and pathological changes in these tissues affect the occurrence and development of OA. Understanding complex crosstalk among different joint tissues and their roles in OA initiation and progression is critical in elucidating the pathogenic mechanism of OA. In this review, we begin with an overview of the role of chondrocytes, synovial cells (synovial fibroblasts and macrophages), mast cells, osteoblasts, osteoclasts, various stem cells, and engineered cells (induced pluripotent stem cells) in OA pathogenesis. Then, we discuss the various mechanisms by which these cells communicate, including paracrine signaling, local microenvironment, co-culture, extracellular vesicles (exosomes), and cell tissue engineering. We particularly focus on the therapeutic potential and clinical applications of stem cell-derived extracellular vesicles, which serve as modulators of cell-to-cell communication, in the field of regenerative medicine, such as cartilage repair. Finally, the challenges and limitations related to exosome-based treatment for OA are discussed. This article provides a comprehensive summary of key cells that might be targets of future therapies for OA.
Collapse
Affiliation(s)
- Zihao Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ziyu Huang
- Foreign Languages College, Shanghai Normal University, Shanghai, China
| | - Lunhao Bai
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
68
|
Oh S, Kwon SH. Extracellular Vesicles in Acute Kidney Injury and Clinical Applications. Int J Mol Sci 2021; 22:8913. [PMID: 34445618 PMCID: PMC8396174 DOI: 10.3390/ijms22168913] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 12/13/2022] Open
Abstract
Acute kidney injury (AKI)--the sudden loss of kidney function due to tissue damage and subsequent progression to chronic kidney disease--has high morbidity and mortality rates and is a serious worldwide clinical problem. Current AKI diagnosis, which relies on measuring serum creatinine levels and urine output, cannot sensitively and promptly report on the state of damage. To address the shortcomings of these traditional diagnosis tools, several molecular biomarkers have been developed to facilitate the identification and ensuing monitoring of AKI. Nanosized membrane-bound extracellular vesicles (EVs) in body fluids have emerged as excellent sources for discovering such biomarkers. Besides this diagnostic purpose, EVs are also being extensively exploited to deliver therapeutic macromolecules to damaged kidney cells to ameliorate AKI. Consequently, many successful AKI biomarker findings and therapeutic applications based on EVs have been made. Here, we review our understanding of how EVs can help with the early identification and accurate monitoring of AKI and be used therapeutically. We will further discuss where current EV-based AKI diagnosis and therapeutic applications fall short and where future innovations could lead us.
Collapse
Affiliation(s)
- Sekyung Oh
- Department of Medical Science, College of Medicine, Catholic Kwandong University, Incheon 22711, Korea;
| | - Sang-Ho Kwon
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
69
|
Agarwal S, Agarwal V, Agarwal M, Singh M. Exosomes: Structure, Biogenesis, Types and Application in Diagnosis and Gene and Drug Delivery. Curr Gene Ther 2021; 20:195-206. [PMID: 32787759 DOI: 10.2174/1566523220999200731011702] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/12/2020] [Accepted: 07/10/2020] [Indexed: 12/21/2022]
Abstract
In recent times, several approaches for targeted gene therapy (GT) had been studied. However, the emergence of extracellular vesicles (EVs) as a shuttle carrying genetic information between cells has gained a lot of interest in scientific communities. Owing to their higher capabilities in dealing with short sequences of nucleic acid (mRNA, miRNA), proteins, recombinant proteins, exosomes, the most popular form of EVs are viewed as reliable biological therapeutic conveyers. They have natural access through every biological membrane and can be employed for site-specific and efficient drug delivery without eliciting any immune responses hence, qualifying as an ideal delivery vehicle. Also, there are many research studies conducted in the last few decades on using exosome-mediated gene therapy into developing an effective therapy with the concept of a higher degree of precision in gene isolation, purification and delivery mechanism loading, delivery and targeting protocols. This review discusses several facets that contribute towards developing an efficient therapeutic regime for gene therapy, highlighting limitations and drawbacks associated with current GT and suggested therapeutic regimes.
Collapse
Affiliation(s)
- Shriya Agarwal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Vinayak Agarwal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Mugdha Agarwal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| | - Manisha Singh
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT) Noida, U.P., India
| |
Collapse
|
70
|
Jayaraman S, Gnanasampanthapandian D, Rajasingh J, Palaniyandi K. Stem Cell-Derived Exosomes Potential Therapeutic Roles in Cardiovascular Diseases. Front Cardiovasc Med 2021; 8:723236. [PMID: 34447796 PMCID: PMC8382889 DOI: 10.3389/fcvm.2021.723236] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
Owing to myocardial abnormalities, cardiac ailments are considered to be the major cause of morbidity and mortality worldwide. According to a recent study, membranous vesicles that are produced naturally, termed as "exosomes", have emerged as the potential candidate in the field of cardiac regenerative medicine. A wide spectrum of stem cells has also been investigated in the treatment of cardiovascular diseases (CVD). Exosomes obtained from the stem cells are found to be cardioprotective and offer great hope in the treatment of CVD. The basic nature of exosomes is to deal with the intracellular delivery of both proteins and nucleic acids. This activity of exosomes helps us to rely on them as the attractive pharmaceutical delivery agents. Most importantly, exosomes derived from microRNAs (miRNAs) hold great promise in assessing the risk of CVD, as they serve as notable biomarkers of the disease. Exosomes are small, less immunogenic, and lack toxicity. These nanovesicles harbor immense potential as a therapeutic entity and would provide fruitful benefits if consequential research were focused on their upbringing and development as a useful diagnostic and therapeutic tool in the field of medicine.
Collapse
Affiliation(s)
- Selvaraj Jayaraman
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Dhanavathy Gnanasampanthapandian
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
| | - Johnson Rajasingh
- Department of Bioscience Research & Medicine-Cardiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Kanagaraj Palaniyandi
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
| |
Collapse
|
71
|
Weiliang Z, Lili G. Research Advances in the Application of Adipose-Derived Stem Cells Derived Exosomes in Cutaneous Wound Healing. Ann Dermatol 2021; 33:309-317. [PMID: 34341631 PMCID: PMC8273313 DOI: 10.5021/ad.2021.33.4.309] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/21/2020] [Accepted: 01/11/2021] [Indexed: 12/15/2022] Open
Abstract
Cutaneous wound healing has always been an intractable medical problem for both clinicians and researchers, with an urgent need for more efficacious methods to achieve optimal outcomes morphologically and functionally. Stem cells, the body's rapid response 'road repair crew,' being on standby to combat tissue injuries, are an essential part of regenerative medicine. Currently, the use of adipose-derived stem cells (ADSCs), a kind of mesenchymal stem cells with multipotent differentiation and self-renewal capacity, is surging in the field of cutaneous wound healing. ADSCs may exert influences either by releasing paracrine signalling factors or differentiating into mature adipose cells to provide the 'building blocks' for engineered tissue. As an important paracrine substance released from ADSCs, exosomes are a kind of extracellular vesicles and carrying various bioactive molecules mediating adjacent or distant intercellular communication. Previous studies have indicated that ADSCs derived exosomes (ADSCs-Exos) promoted skin wound healing by affecting all stages of wound healing, including regulating inflammatory response, promoting proliferation and migration of fibroblasts or keratinocytes, facilitating angiogenesis, and regulating remodeling of extracellular matrix, which have provided new opportunities for understanding how ADSCs-Exos mediate intercellular communication in pathological processes of the skin and therapeutic strategies for cutaneous wound repair. In this review, we focus on elucidating the role of ADSCs-Exos at various stages of cutaneous wound healing, detailing the latest developments, and presenting some challenges necessary to be addressed in this field, with the expectation of providing a new perspective on how to best utilize this powerful cell-free therapy in the future.
Collapse
Affiliation(s)
- Zeng Weiliang
- Department of Cosmetic and Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guo Lili
- Department of Cosmetic and Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
72
|
Escudé Martinez de Castilla P, Tong L, Huang C, Sofias AM, Pastorin G, Chen X, Storm G, Schiffelers RM, Wang JW. Extracellular vesicles as a drug delivery system: A systematic review of preclinical studies. Adv Drug Deliv Rev 2021; 175:113801. [PMID: 34015418 DOI: 10.1016/j.addr.2021.05.011] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/10/2021] [Accepted: 05/15/2021] [Indexed: 02/06/2023]
Abstract
During the past decades, extracellular vesicles (EVs) have emerged as an attractive drug delivery system. Here, we assess their pre-clinical applications, in the form of a systematic review. For each study published in the past decade, disease models, animal species, EV donor cell types, active pharmaceutical ingredients (APIs), EV surface modifications, API loading methods, EV size and charge, estimation of EV purity, presence of biodistribution studies and administration routes were quantitatively analyzed in a defined and reproducible way. We have interpreted the trends we observe over the past decade, to define the niches where to apply EVs for drug delivery in the future and to provide a basis for regulatory guidelines.
Collapse
|
73
|
Ghafouri-Fard S, Niazi V, Hussen BM, Omrani MD, Taheri M, Basiri A. The Emerging Role of Exosomes in the Treatment of Human Disorders With a Special Focus on Mesenchymal Stem Cells-Derived Exosomes. Front Cell Dev Biol 2021; 9:653296. [PMID: 34307345 PMCID: PMC8293617 DOI: 10.3389/fcell.2021.653296] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are produced by diverse eukaryotic and prokaryotic cells. They have prominent roles in the modulation of cell-cell communication, inflammation versus immunomodulation, carcinogenic processes, cell proliferation and differentiation, and tissue regeneration. These acellular vesicles are more promising than cellular methods because of the lower risk of tumor formation, autoimmune responses and toxic effects compared with cell therapy. Moreover, the small size and lower complexity of these vesicles compared with cells have made their production and storage easier than cellular methods. Exosomes originated from mesenchymal stem cells has also been introduced as therapeutic option for a number of human diseases. The current review aims at summarization of the role of EVs in the regenerative medicine with a focus on their therapeutic impacts in liver fibrosis, lung disorders, osteoarthritis, colitis, myocardial injury, spinal cord injury and retinal injury.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Niazi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Mir Davood Omrani
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
74
|
Pan W, Xu X, Zhang M, Song X. Human urine-derived stem cell-derived exosomal miR-21-5p promotes neurogenesis to attenuate Rett syndrome via the EPha4/TEK axis. J Transl Med 2021; 101:824-836. [PMID: 33976355 DOI: 10.1038/s41374-021-00574-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 11/09/2022] Open
Abstract
Rett syndrome (RTT) is a rare neurodevelopmental disorder that results in multiple disabilities. Exosomal microRNA (miRs) from urine-derived stem cells (USCs) have been shown to induce neurogenesis and aid in functional recovery from brain ischemia. In the present study, we sought to determine whether that exosomal miR-21-5p from USCs could promote early neural formation in a model of RTT. USCs were isolated and evaluated by flow cytometry. Exosomes were analyzed by transmission electron microscopy, tunable resistive pulse sensing (TRPS), and western blotting. PKH26 fluorescent dyes were used to observe intake of exosomes in vivo and in vitro. An RTT mouse model was treated with exosomes for behavioral studies. Dual-luciferase report gene assays were conducted to evaluate the relationship between miR-21-5p and Eph receptor A4 (EphA4). In vitro, treatment with exosomes from human urine-derived stem cells (USC-Exos) increased the percentage of neuron-specific class III beta-tubulin (Tuj1)+ nerve cells as well as the transcription levels of β-III tubulin and doublecortin (DCX). A higher level of miR-21-5p was observed in USC-Exos, which promoted differentiation in NSCs by targeting the EPha4/TEK axis. In vivo, exosomal miR-21-5p improved the behavior, motor coordination, and cognitive ability of mice, facilitated the differentiation of NSCs in the subventricular zone of the lateral ventricle and promoted a marked rise in the number of DCX+ cells. Our data provide evidence that exosomal miR-21-5p from human USCs facilitate early nerve formation by regulating the EPha4/TEK axis.
Collapse
Affiliation(s)
- Wei Pan
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Xiaoheng Xu
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Meng Zhang
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Xingyu Song
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, People's Republic of China.
| |
Collapse
|
75
|
Wei Z, Chen Z, Zhao Y, Fan F, Xiong W, Song S, Yin Y, Hu J, Yang K, Yang L, Xu B, Ge J. Mononuclear phagocyte system blockade using extracellular vesicles modified with CD47 on membrane surface for myocardial infarction reperfusion injury treatment. Biomaterials 2021; 275:121000. [PMID: 34218049 DOI: 10.1016/j.biomaterials.2021.121000] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/24/2021] [Accepted: 06/26/2021] [Indexed: 12/16/2022]
Abstract
Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) with anti-apoptotic and anti-inflammatory properties have been intensively studied. However, rapid clearance by the mononuclear phagocyte system remains a huge barrier for the delivery of extracellular vesicle contents into target organs and restricts its wider application, particularly in the heart. CD47 is a transmembrane protein that enables cancer cells to evade clearance by macrophages through CD47- signal regulatory proteinα binding, which initiates a "don't eat me" signal. This study aimed to explore the biodistribution and delivery efficiency of EVs carrying the membrane protein CD47 and specific anti-apoptotic miRNAs. EVs were isolated from MSCs overexpressing CD47 (CD47-EVs) and identified. Fluorescence-labeled EVs were injected through the tail vein and tracked using fluorescence imaging. In silico analysis was performed to determine miRNA profiles in MSCs and in a heart-derived H9c2 cardiomyoblast cell line under hypoxia vs. normoxia conditions. Electro CD47-EV was constructed by encapsulating purified CD47-EV with miR-21a via electroporation. The effect of miR21-EVs on the pro-apoptotic gene encoding phosphatase and tensin homolog (PTEN) was evaluated by dual-luciferase assay, qPCR, and western blotting. Exogenous miR21 distribution, PTEN protein level, blood vessel density, anti-apoptotic effect by TdT-mediated dUTP nick-end labeling staining, and macrophage and leukocyte infiltration in the myocardium were assessed by immunofluorescence staining. Cardiac functional recovery during the early stage and recovery period was evaluated using echocardiography. The results showed that CD47-EVs were still detectable in the plasma 120 min after the tail vein injection, compared to the detection time of less than 30 min observed with the unmodified EVs. More strikingly, CD47-EVs preferentially accumulated in the heart in the ischemia-reperfusion (I/R) + CD47-EV group [heart total fluorescence radiance ( × 105 Photons/sec/cm2/sr) 51.62 ± 11.30 v.s. 10.08 ± 3.15 in the I/R + unmodified EVs group] 8 h post-injection. Exogenous miR-21 is efficiently internalized into cardiomyocytes, inhibits apoptosis, alleviates inflammation, and improves cardiac function. In conclusion, electro CD47-EVs efficiently improve biodistribution in the heart, shedding new light on the application of a two-step EV delivery method (CD47 genetic modification followed by therapeutic content electrotransfection) as a potential therapeutic tool for myocardial I/R injury that may benefit patients in the future.
Collapse
Affiliation(s)
- Zilun Wei
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Biomedical Science, Fudan University, Shanghai, China; Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Zhaoyang Chen
- Cardiology Department, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yongchao Zhao
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Biomedical Science, Fudan University, Shanghai, China; Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Fan Fan
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Weidong Xiong
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Biomedical Science, Fudan University, Shanghai, China; Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Shuai Song
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Yong Yin
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Jingjing Hu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Kun Yang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Lebing Yang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China; Department of Cardiology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, China.
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Biomedical Science, Fudan University, Shanghai, China.
| |
Collapse
|
76
|
Malik S, Sundarrajan S, Hussain T, Nazir A, Ramakrishna S. Fabrication of Highly Oriented Cylindrical Polyacrylonitrile, Poly(lactide- co-glycolide), Polycaprolactone and Poly(vinyl acetate) Nanofibers for Vascular Graft Applications. Polymers (Basel) 2021; 13:2075. [PMID: 34202499 PMCID: PMC8271820 DOI: 10.3390/polym13132075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 11/23/2022] Open
Abstract
Small-diameter vascular grafts fabricated from synthetic polymers have found limited applications so far in vascular surgeries, owing to their poor mechanical properties. In this study, cylindrical nanofibrous structures of highly oriented nanofibers made from polyacrylonitrile, poly (lactide-co-glycolide) (PLGA), polycaprolactone (PCL) and poly(vinyl acetate) (PVAc) were investigated. Cylindrical collectors with alternate conductive and non-conductive segments were used to obtain highly oriented nanofibrous structures at the same time with better mechanical properties. The surface morphology (orientation), mechanical properties and suture retention of the nanofibrous structures were characterized using SEM, mechanical tester and universal testing machine, respectively. The PLGA nanofibrous cylindrical structure exhibited excellent properties (tensile strength of 9.1 ± 0.6 MPa, suture retention strength of 27N and burst pressure of 350 ± 50 mmHg) when compared to other polymers. Moreover, the PLGA grafts showed good porosity and elongation values, that could be potentially used for vascular graft applications. The combination of PLGA nanofibers with extracellular vesicles (EVs) will be explored as a potential vascular graft in future.
Collapse
Affiliation(s)
- Sairish Malik
- Electrospun Materials & Polymeric Membranes Research Group (EMPMRG), National Textile University, Sheikhupura Road, Faisalabad 37610, Pakistan; (S.M.); (T.H.); (A.N.)
| | - Subramanian Sundarrajan
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore;
| | - Tanveer Hussain
- Electrospun Materials & Polymeric Membranes Research Group (EMPMRG), National Textile University, Sheikhupura Road, Faisalabad 37610, Pakistan; (S.M.); (T.H.); (A.N.)
| | - Ahsan Nazir
- Electrospun Materials & Polymeric Membranes Research Group (EMPMRG), National Textile University, Sheikhupura Road, Faisalabad 37610, Pakistan; (S.M.); (T.H.); (A.N.)
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore;
| |
Collapse
|
77
|
Ćulum NM, Cooper TT, Bell GI, Hess DA, Lagugné-Labarthet F. Characterization of extracellular vesicles derived from mesenchymal stromal cells by surface-enhanced Raman spectroscopy. Anal Bioanal Chem 2021; 413:5013-5024. [PMID: 34137912 DOI: 10.1007/s00216-021-03464-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 12/18/2022]
Abstract
Extracellular vesicles (EVs) are secreted by all cells into bodily fluids and play an important role in intercellular communication through the transfer of proteins and RNA. There is evidence that EVs specifically released from mesenchymal stromal cells (MSCs) are potent cell-free regenerative agents. However, for MSC EVs to be used in therapeutic practices, there must be a standardized and reproducible method for their characterization. The detection and characterization of EVs are a challenge due to their nanoscale size as well as their molecular heterogeneity. To address this challenge, we have fabricated gold nanohole arrays of varying sizes and shapes by electron beam lithography. These platforms have the dual purpose of trapping single EVs and enhancing their vibrational signature in surface-enhanced Raman spectroscopy (SERS). In this paper, we report SERS spectra for MSC EVs derived from pancreatic tissue (Panc-MSC) and bone marrow (BM-MSC). Using principal component analysis (PCA), we determined that the main compositional differences between these two groups are found at 1236, 761, and 1528 cm-1, corresponding to amide III, tryptophan, and an in-plane -C=C- vibration, respectively. We additionally explored several machine learning approaches to distinguish between BM- and Panc-MSC EVs and achieved 89 % accuracy, 89 % sensitivity, and 88 % specificity using logistic regression.
Collapse
Affiliation(s)
- Nina M Ćulum
- Department of Chemistry, Centre for Advanced Materials and Biomaterials Research (CAMBR), University of Western Ontario (Western University), 1151 Richmond St, London, Ontario, N6A 5B7, Canada
| | - Tyler T Cooper
- Robarts Research Institute, Department of Physiology and Pharmacology, Schulich School of medicine and Dentistry, University of Western Ontario (Western University), 1151 Richmond St, London, Ontario, N6A 5B7, Canada
| | - Gillian I Bell
- Robarts Research Institute, Department of Physiology and Pharmacology, Schulich School of medicine and Dentistry, University of Western Ontario (Western University), 1151 Richmond St, London, Ontario, N6A 5B7, Canada
| | - David A Hess
- Robarts Research Institute, Department of Physiology and Pharmacology, Schulich School of medicine and Dentistry, University of Western Ontario (Western University), 1151 Richmond St, London, Ontario, N6A 5B7, Canada
| | - François Lagugné-Labarthet
- Department of Chemistry, Centre for Advanced Materials and Biomaterials Research (CAMBR), University of Western Ontario (Western University), 1151 Richmond St, London, Ontario, N6A 5B7, Canada.
| |
Collapse
|
78
|
Endothelial Progenitor Cell-Derived Extracellular Vesicles: Potential Therapeutic Application in Tissue Repair and Regeneration. Int J Mol Sci 2021; 22:ijms22126375. [PMID: 34203627 PMCID: PMC8232313 DOI: 10.3390/ijms22126375] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/01/2021] [Accepted: 06/11/2021] [Indexed: 12/19/2022] Open
Abstract
Recently, many studies investigated the role of a specific type of stem cell named the endothelial progenitor cell (EPC) in tissue regeneration and repair. EPCs represent a heterogeneous population of mononuclear cells resident in the adult bone marrow. EPCs can migrate and differentiate in injured sites or act in a paracrine way. Among the EPCs’ secretome, extracellular vesicles (EVs) gained relevance due to their possible use for cell-free biological therapy. They are more biocompatible, less immunogenic, and present a lower oncological risk compared to cell-based options. EVs can efficiently pass the pulmonary filter and deliver to target tissues different molecules, such as micro-RNA, growth factors, cytokines, chemokines, and non-coding RNAs. Their effects are often analogous to their cellular counterparts, and EPC-derived EVs have been tested in vitro and on animal models to treat several medical conditions, including ischemic stroke, myocardial infarction, diabetes, and acute kidney injury. EPC-derived EVs have also been studied for bone, brain, and lung regeneration and as carriers for drug delivery. This review will discuss the pre-clinical evidence regarding EPC-derived EVs in the different disease models and regenerative settings. Moreover, we will discuss the translation of their use into clinical practice and the possible limitations of this process.
Collapse
|
79
|
Yao Z, Li J, Xiong H, Cui H, Ning J, Wang S, Ouyang X, Qian Y, Fan C. MicroRNA engineered umbilical cord stem cell-derived exosomes direct tendon regeneration by mTOR signaling. J Nanobiotechnology 2021; 19:169. [PMID: 34090456 PMCID: PMC8180131 DOI: 10.1186/s12951-021-00906-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/21/2021] [Indexed: 12/27/2022] Open
Abstract
Background Exosomes are extracellular vesicles of nano-structures and represent an emerging nano-scale acellular therapy in recent years. Tendon regeneration is a sophisticated process in the field of microsurgery due to its poor natural healing ability. To date, no successful long-term solution has been provided for the healing of tendon injuries. Functional recovery requires advanced treatment strategies. Human umbilical cord mesenchymal stem cell-derived exosomes (HUMSC-Exos) are considered as promising cell-free therapeutic agents. However, few studies reported their potential in the tendon repair previously. In this study, we explored the roles and underlying mechanisms of HUMSC-Exos in the tendon regeneration. Results Expression of tendon‐specific markers in, and collagen deposition by, tendon-derived stem cells (TDSCs) treated with HUMSC-Exos increased in vitro. In a rat Achilles tendon injury model, treatment with HUMSC-Exos improved the histological structure, enhanced tendon-specific matrix components, and optimized biomechanical properties of the Achilles tendon. Findings in miRNA sequencing indicated a significant increase in miR-29a-3p in HUMSC-Exo-treated Achilles tendons. Next, luciferase assay in combination with western blot identified phosphatase and tensin homolog (PTEN) as the specific target of miR-29a-3p. Furthermore, we applied a miR-29a-3p-specific agonist to engineer HUMSC-Exos. These HUMSC-Exos overexpressing miR-29a-3p amplified the gain effects of HUMSC-Exos on tendon healing in vivo. To explore the underlying mechanisms, a transforming growth factor-β1 (TGF-β1) inhibitor (SB-431542), mTOR inhibitor (rapamycin), and engineered HUMSC-Exos were employed. The results showed that TGF-β1 and mTOR signaling were involved in the beneficial effects of HUMSC-Exos on tendon regeneration. Conclusion The findings in our study suggest that PTEN/mTOR/TGF-β1 signaling cascades may be a potential pathway for HUMSC-Exos to deliver miR-29a-3p for tendon healing and implicate a novel therapeutic strategy for tendon regeneration via engineered stem cell-derived exosomes. Graphic abstract ![]()
Collapse
Affiliation(s)
- Zhixiao Yao
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Juehong Li
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Hao Xiong
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Haomin Cui
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Jiexin Ning
- Department of Plastics, Binzhou People's Hospital, Binzhou, 256610, China
| | - Shikun Wang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xingyu Ouyang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yun Qian
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
80
|
Xia LZ, Tao J, Chen YJ, Liang LL, Luo GF, Cai ZM, Wang Z. Factors Affecting the Re-Endothelialization of Endothelial Progenitor Cell. DNA Cell Biol 2021; 40:1009-1025. [PMID: 34061680 DOI: 10.1089/dna.2021.0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The vascular endothelium, which plays an essential role in maintaining the normal shape and function of blood vessels, is a natural barrier between the circulating blood and the vascular wall tissue. The endothelial damage can cause vascular lesions, such as atherosclerosis and restenosis. After the vascular intima injury, the body starts the endothelial repair (re-endothelialization) to inhibit the neointimal hyperplasia. Endothelial progenitor cell is the precursor of endothelial cells and plays an important role in the vascular re-endothelialization. However, re-endothelialization is inevitably affected in vivo and in vitro by factors, which can be divided into two types, namely, promotion and inhibition, and act on different links of the vascular re-endothelialization. This article reviews these factors and related mechanisms.
Collapse
Affiliation(s)
- Lin-Zhen Xia
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Jun Tao
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Yan-Jun Chen
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Ling-Li Liang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Gui-Fang Luo
- Department of Gynaecology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Ze-Min Cai
- Pediatrics Department, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Zuo Wang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
81
|
Hamzah RN, Alghazali KM, Biris AS, Griffin RJ. Exosome Traceability and Cell Source Dependence on Composition and Cell-Cell Cross Talk. Int J Mol Sci 2021; 22:5346. [PMID: 34069542 PMCID: PMC8161017 DOI: 10.3390/ijms22105346] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are small vesicles with an average diameter of 100 nm that are produced by many, if not all, cell types. Exosome cargo includes lipids, proteins, and nucleic acids arranged specifically in the endosomes of donor cells. Exosomes can transfer the donor cell components to target cells and can affect cell signaling, proliferation, and differentiation. Important new information about exosomes' remote communication with other cells is rapidly being accumulated. Recent data indicates that the results of this communication depend on the donor cell type and the environment of the host cell. In the field of cancer research, major questions remain, such as whether tumor cell exosomes are equally taken up by cancer cells and normal cells and whether exosomes secreted by normal cells are specifically taken up by other normal cells or also tumor cells. Furthermore, we do not know how exosome uptake is made selective, how we can trace exosome uptake selectivity, or what the most appropriate methods are to study exosome uptake and selectivity. This review will explain the effect of exosome source and the impact of the donor cell growth environment on tumor and normal cell interaction and communication. The review will also summarize the methods that have been used to label and trace exosomes to date.
Collapse
Affiliation(s)
- Rabab N. Hamzah
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR 72204, USA; (K.M.A.); (A.S.B.)
| | - Karrer M. Alghazali
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR 72204, USA; (K.M.A.); (A.S.B.)
- Nushores Biosciences LLC, Little Rock, AR 72211, USA
| | - Alexandru S. Biris
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR 72204, USA; (K.M.A.); (A.S.B.)
| | - Robert J. Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| |
Collapse
|
82
|
Zhou Y, Zhao B, Zhang XL, Lu YJ, Lu ST, Cheng J, Fu Y, Lin L, Zhang NY, Li PX, Zhang J, Zhang J. Combined topical and systemic administration with human adipose-derived mesenchymal stem cells (hADSC) and hADSC-derived exosomes markedly promoted cutaneous wound healing and regeneration. Stem Cell Res Ther 2021; 12:257. [PMID: 33933157 PMCID: PMC8088044 DOI: 10.1186/s13287-021-02287-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
Background Cutaneous wound healing and regeneration have become a recognized health challenge in the world, which causes severe damage to the mental and physical health of patients. Human adipose-derived mesenchymal stem cells (hADSC) play an essential role in wound healing via their paracrine function. Exosomes secreted by hADSC may contribute to this progress. In this study, we investigated the potential clinical application roles of hADSC and hADSC-derived exosomes (hADSC-Exo) in cutaneous wound healing. Methods hADSC-Exo was isolated from human hADSC by ultracentrifugation. Mice were subjected to a full-thickness skin biopsy experiment and treated with either control vehicle or hADSC or hADSC-Exo by smearing administration (sm) or subcutaneous administration (sc) or intravenous administration (iv). The efficacy of hADSC and hADSC-Exo on wound healing was evaluated by measuring wound closure rates, histological analysis. Results Combined application of local hADSC-Exo smearing and hADSC/hADSC-Exo intravenous administration offered the additional benefit of promoting wound healing, accelerating re-epithelialization, reducing scar widths, and enhancing angiogenesis and collagen synthesis. Either topical application of hADSC-Exo or systemic administration with hADSC/hADSC-Exo appeared more effective in stimulating cell proliferation, inhibiting cell apoptosis and inflammation, and promoting skin elasticity and barrier integrity, with increased genes expression of PCNA, VEGF, collagen III, Filaggrin, Loricrin, and AQP3, with decreased genes expression of TNF-alpha. Conclusion Our findings suggest that the combined administration of hADSC/hADSC-Exo can facilitate cutaneous wound healing and reduce scar formation. These data provide the first evidence for the feasibility of smearing of hADSC-Exo as a cell-free therapy in treating cutaneous wounds, and the potential clinical value of combined administration of hADSC/hADSC-Exo. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02287-9.
Collapse
Affiliation(s)
- Yang Zhou
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Bo Zhao
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Stem Cell Translational Research Center of Tongji Hospital, School of Life Science and Technology, Tongji University, 389 Xincun Road, Shanghai, 200065, China
| | - Xin-Liao Zhang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yi-Jun Lu
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Shou-Tao Lu
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Jian Cheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Stem Cell Translational Research Center of Tongji Hospital, School of Life Science and Technology, Tongji University, 389 Xincun Road, Shanghai, 200065, China
| | - Yu Fu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Stem Cell Translational Research Center of Tongji Hospital, School of Life Science and Technology, Tongji University, 389 Xincun Road, Shanghai, 200065, China
| | - Lin Lin
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Ning-Yan Zhang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Stem Cell Translational Research Center of Tongji Hospital, School of Life Science and Technology, Tongji University, 389 Xincun Road, Shanghai, 200065, China
| | - Pei-Xin Li
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Jing Zhang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Stem Cell Translational Research Center of Tongji Hospital, School of Life Science and Technology, Tongji University, 389 Xincun Road, Shanghai, 200065, China. .,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China. .,Tongji Lifeng Institute of Regenerative Medicine, Tongji University, Shanghai, China.
| | - Jun Zhang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China. .,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China. .,Tongji Lifeng Institute of Regenerative Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
83
|
Li Q, Yu H, Sun M, Yang P, Hu X, Ao Y, Cheng J. The tissue origin effect of extracellular vesicles on cartilage and bone regeneration. Acta Biomater 2021; 125:253-266. [PMID: 33657452 DOI: 10.1016/j.actbio.2021.02.039] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/22/2021] [Accepted: 02/24/2021] [Indexed: 12/30/2022]
Abstract
Direct implantation of mesenchymal stem cells (MSCs) for cartilage and bone tissue engineering faces challenges, such as immune rejection and loss of cellular viability or functionality. As nanoscale natural particles, exosomes or small extracellular vesicles (EVs) of MSCs have potential to circumvent these problems. It is significant to investigate the impact of the tissue origin of MSCs on the therapeutic bioactivity of their corresponding EVs for cartilage and bone regeneration. Here, rat MSCs isolated from the adipose, bone marrow, and synovium are cultured to obtain their corresponding EVs (ADSC-EVs, BMSC-EVs, and SMSC-EVs, respectively). The ADSC-EVs stimulate the migration, proliferation, and chondrogenic and osteogenic differentiation of BMSCs in vitro as well as cartilage and bone regeneration in a mouse model more than the BMSC-EVs or SMSC-EVs. Proteomics analysis reveals that the tissue origin contributes to the distinct protein profiles among the three types of EVs, which induced cartilage and bone regenerative capacities by potential mechanisms of regulating signaling pathways including focal adhesion, ECM-receptor interaction, actin cytoskeleton, cAMP, and PI3K-Akt signaling pathways. Consequently, these findings provide insight that the adipose may be a superior candidate in EV-based nanomedicine for cartilage and bone regeneration. STATEMENT OF SIGNIFICANCE: Extracelluar vesicles (EVs) of mesenchymal stem cells (MSCs) have been considered as a promising approach in cartilage and bone tissue engineering. In this study, for the first time, we investigated the tissue origin effect of EVs on chondrogenesis and osteogenesis of MSCs in vitro and in vivo. The results demonstrated that EVs of adipose-derived MSCs showed the most efficiency. Meanwhile, protein proteomics revealed the potential mechanisms. We provide a novel evidence that the adipose is a superior reservoir in EV-based nanotechnologies and biomaterials for cartilage and bone regeneration.
Collapse
Affiliation(s)
- Qi Li
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Huilei Yu
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Muyang Sun
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Peng Yang
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Xiaoqing Hu
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China
| | - Yingfang Ao
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China.
| | - Jin Cheng
- Department of Sports Medicine, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China.
| |
Collapse
|
84
|
Imanishi Y, Hata M, Matsukawa R, Aoyagi A, Omi M, Mizutani M, Naruse K, Ozawa S, Honda M, Matsubara T, Takebe J. Efficacy of extracellular vesicles from dental pulp stem cells for bone regeneration in rat calvarial bone defects. Inflamm Regen 2021; 41:12. [PMID: 33853679 PMCID: PMC8048358 DOI: 10.1186/s41232-021-00163-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/30/2021] [Indexed: 12/27/2022] Open
Abstract
Background Extracellular vesicles (EVs) are known to be secreted by various cells. In particular, mesenchymal stem cell (MSC)-derived EVs (MSC-EVs) have tissue repair capacity and anti-inflammatory properties. Dental pulp stem cells (DPSCs), which are MSCs isolated from pulp tissue, are less invasive to the body than other MSCs and can be collected from young individuals. In this study, we investigated the efficacy of EVs secreted by DPSCs (DPSC-EVs) for bone formation. Methods DPSC-EVs were isolated from the cell culture medium of DPSCs. DPSC-EVs were unilaterally injected along with collagen (COL), beta-tricalcium phosphate (β-TCP) or hydroxyapatite (HA) into rat calvarial bone defects. The effects of DPSC-EVs were analyzed by micro-computed tomography (micro-CT) and histological observation. Results Micro-CT showed that administration of DPSC-EVs with the abovementioned scaffolds resulted in bone formation in the periphery of the defects. DPSC-EVs/COL specifically resulted in bone formation in the center of the defects. Histological observation revealed that DPSC-EVs/COL promoted new bone formation. Administration of DPSC-EVs/COL had almost the same effect on the bone defect site as transplantation of DPSCs/COL. Conclusions These results suggest that DPSC-EVs may be effective tools for bone tissue regeneration.
Collapse
Affiliation(s)
- Yuka Imanishi
- Department of Removable Prosthodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, 464-8651, Japan
| | - Masaki Hata
- Department of Removable Prosthodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, 464-8651, Japan.
| | - Ryohei Matsukawa
- Department of Removable Prosthodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, 464-8651, Japan
| | - Atsushi Aoyagi
- Department of Removable Prosthodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, 464-8651, Japan
| | - Maiko Omi
- Department of Removable Prosthodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, 464-8651, Japan
| | - Makoto Mizutani
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Keiko Naruse
- Department of Internal Medicine, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Shogo Ozawa
- Department of Removable Prosthodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, 464-8651, Japan
| | - Masaki Honda
- Department of Oral Anatomy, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Tatsuaki Matsubara
- Department of Internal Medicine, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Jun Takebe
- Department of Removable Prosthodontics, School of Dentistry, Aichi Gakuin University, 2-11 Suemori-dori, Chikusa-ku, Nagoya, 464-8651, Japan
| |
Collapse
|
85
|
Maiullari F, Chirivì M, Costantini M, Ferretti AM, Recchia S, Maiullari S, Milan M, Presutti D, Pace V, Raspa M, Scavizzi F, Massetti M, Petrella L, Fanelli M, Rizzi M, Fortunato O, Moretti F, Caradonna E, Bearzi C, Rizzi R. In vivoorganized neovascularization induced by 3D bioprinted endothelial-derived extracellular vesicles. Biofabrication 2021; 13. [PMID: 33434889 DOI: 10.1088/1758-5090/abdacf] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/12/2021] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs) have become a key tool in the biotechnological landscape due to their well-documented ability to mediate intercellular communication. This feature has been explored and is under constant investigation by researchers, who have demonstrated the important role of EVs in several research fields ranging from oncology to immunology and diagnostics to regenerative medicine. Unfortunately, there are still some limitations to overcome before clinical application, including the inability to confine the EVs to strategically defined sites of interest to avoid side effects. In this study, for the first time, EV application is supported by 3D bioprinting technology to develop a new strategy for applying the angiogenic cargo of human umbilical vein endothelial cell-derived EVs in regenerative medicine. EVs, derived from human endothelial cells and grown under different stressed conditions, were collected and used as bioadditives for the formulation of advanced bioinks. Afterin vivosubcutaneous implantation, we demonstrated that the bioprinted 3D structures, loaded with EVs, supported the formation of a new functional vasculaturein situ, consisting of blood-perfused microvessels recapitulating the printed pattern. The results obtained in this study favour the development of new therapeutic approaches for critical clinical conditions, such as the need for prompt revascularization of ischaemic tissues, which represent the fundamental substrate for advanced regenerative medicine applications.
Collapse
Affiliation(s)
- Fabio Maiullari
- Gemelli Molise SpA, Campobasso, Italy.,Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', Milan, Italy
| | - Maila Chirivì
- Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', Milan, Italy.,Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy
| | - Marco Costantini
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Maria Ferretti
- Institute of Chemical Sciences and Technologies "Giulio Natta", National Research Council of Italy (SCITEC-CNR), Milano, Italy
| | - Sandro Recchia
- Department of Science and High Technology, University of Insubria, Como, Italy
| | - Silvia Maiullari
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy.,Institute of Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marika Milan
- Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', Milan, Italy.,Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy
| | - Dario Presutti
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy.,Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Valentina Pace
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy.,Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Marcello Raspa
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy
| | - Ferdinando Scavizzi
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy
| | - Massimo Massetti
- Department of Cardiovascular Disease, IRCCS Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Lella Petrella
- Laboratory of Molecular Oncology, Gemelli Molise SpA, Campobasso, Italy
| | - Mara Fanelli
- Laboratory of Molecular Oncology, Gemelli Molise SpA, Campobasso, Italy
| | - Marta Rizzi
- Ufficio Programmazione e Grant Office, National Research Council of Italy (UPGO-CNR), Rome, Italy
| | - Orazio Fortunato
- Tumor Genomics Unit, Department of Research, IRCCS Fondazione Istituto Nazionale dei Tumori, Milan, Italy
| | - Fabiola Moretti
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy.,IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Claudia Bearzi
- Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', Milan, Italy.,Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy
| | - Roberto Rizzi
- Istituto Nazionale Genetica Molecolare INGM 'Romeo ed Enrica Invernizzi', Milan, Italy.,Institute of Biomedical Technologies, National Research Council of Italy (ITB-CNR), Segrate, Milan, Italy
| |
Collapse
|
86
|
Advances and Perspectives in Dental Pulp Stem Cell Based Neuroregeneration Therapies. Int J Mol Sci 2021; 22:ijms22073546. [PMID: 33805573 PMCID: PMC8036729 DOI: 10.3390/ijms22073546] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Human dental pulp stem cells (hDPSCs) are some of the most promising stem cell types for regenerative therapies given their ability to grow in the absence of serum and their realistic possibility to be used in autologous grafts. In this review, we describe the particular advantages of hDPSCs for neuroregenerative cell therapies. We thoroughly discuss the knowledge about their embryonic origin and characteristics of their postnatal niche, as well as the current status of cell culture protocols to maximize their multilineage differentiation potential, highlighting some common issues when assessing neuronal differentiation fates of hDPSCs. We also review the recent progress on neuroprotective and immunomodulatory capacity of hDPSCs and their secreted extracellular vesicles, as well as their combination with scaffold materials to improve their functional integration on the injured central nervous system (CNS) and peripheral nervous system (PNS). Finally, we offer some perspectives on the current and possible future applications of hDPSCs in neuroregenerative cell therapies.
Collapse
|
87
|
Laggner M, Gugerell A, Copic D, Jeitler M, Springer M, Peterbauer A, Kremslehner C, Filzwieser-Narzt M, Gruber F, Madlener S, Erb M, Widder J, Lechner W, Georg D, Mildner M, Ankersmit HJ. Comparing the efficacy of γ- and electron-irradiation of PBMCs to promote secretion of paracrine, regenerative factors. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:14-27. [PMID: 33768126 PMCID: PMC7960502 DOI: 10.1016/j.omtm.2021.02.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 02/19/2021] [Indexed: 11/28/2022]
Abstract
Cell-free secretomes represent a promising new therapeutic avenue in regenerative medicine, and γ-irradiation of human peripheral blood mononuclear cells (PBMCs) has been shown to promote the release of paracrine factors with high regenerative potential. Recently, the use of alternative irradiation sources, such as artificially generated β- or electron-irradiation, is encouraged by authorities. Since the effect of the less hazardous electron-radiation on the production and functions of paracrine factors has not been tested so far, we compared the effects of γ- and electron-irradiation on PBMCs and determined the efficacy of both radiation sources for producing regenerative secretomes. Exposure to 60 Gy γ-rays from a radioactive nuclide and 60 Gy electron-irradiation provided by a linear accelerator comparably induced cell death and DNA damage. The transcriptional landscapes of PBMCs exposed to either radiation source shared a high degree of similarity. Secretion patterns of proteins, lipids, and extracellular vesicles displayed similar profiles after γ- and electron-irradiation. Lastly, we detected comparable biological activities in functional assays reflecting the regenerative potential of the secretomes. Taken together, we were able to demonstrate that electron-irradiation is an effective, alternative radiation source for producing therapeutic, cell-free secretomes. Our study paves the way for future clinical trials employing secretomes generated with electron-irradiation in tissue-regenerative medicine.
Collapse
Affiliation(s)
- Maria Laggner
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria.,Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090 Vienna, Austria
| | - Alfred Gugerell
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria.,Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090 Vienna, Austria
| | - Dragan Copic
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria.,Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090 Vienna, Austria
| | - Markus Jeitler
- Core Facility Genomics, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Springer
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria.,Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090 Vienna, Austria
| | - Anja Peterbauer
- Austrian Red Cross Blood Transfusion Service of Upper Austria, 4020 Linz, Austria
| | - Christopher Kremslehner
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria.,Christian Doppler Laboratory for Biotechnology of Skin Aging, 1090 Vienna, Austria
| | - Manuel Filzwieser-Narzt
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria.,Christian Doppler Laboratory for Biotechnology of Skin Aging, 1090 Vienna, Austria
| | - Florian Gruber
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria.,Christian Doppler Laboratory for Biotechnology of Skin Aging, 1090 Vienna, Austria
| | - Sibylle Madlener
- Molecular Neuro-Oncology, Department of Pediatrics and Adolescent Medicine, and Institute of Neurology, Medical University of Vienna, 1090 Vienna, Austria.,Comprehensive Cancer Center of the Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Erb
- SYNLAB Analytics and Services Switzerland AG, 4127 Birsfelden, Switzerland
| | - Joachim Widder
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Wolfgang Lechner
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Dietmar Georg
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Hendrik Jan Ankersmit
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria.,Laboratory for Cardiac and Thoracic Diagnosis and Regeneration, 1090 Vienna, Austria
| |
Collapse
|
88
|
A bioactive injectable self-healing anti-inflammatory hydrogel with ultralong extracellular vesicles release synergistically enhances motor functional recovery of spinal cord injury. Bioact Mater 2021; 6:2523-2534. [PMID: 33615043 PMCID: PMC7873581 DOI: 10.1016/j.bioactmat.2021.01.029] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/17/2021] [Accepted: 01/24/2021] [Indexed: 12/19/2022] Open
Abstract
The repair and motor functional recovery after spinal cord injury (SCI) remains a worldwide challenge. The inflammatory microenvironment is one of main obstacles on inhibiting the recovery of SCI. Using mesenchymal stem cells (MSCs) derived extracellular vesicles to replace MSCs transplantation and mimic cell paracrine secretions provides a potential strategy for microenvironment regulation. However, the effective preservation and controlled release of extracellular vesicles in the injured spinal cord tissue are still not satisfied. Herein, we fabricated an injectable adhesive anti-inflammatory F127-polycitrate-polyethyleneimine hydrogel (FE) with sustainable and long term extracellular vesicle release (FE@EVs) for improving motor functional recovery after SCI. The orthotopic injection of FE@EVs hydrogel could encapsulate extracellular vesicles on the injured spinal cord, thereby synergistically induce efficient integrated regulation through suppressing fibrotic scar formation, reducing inflammatory reaction, promoting remyelination and axonal regeneration. This study showed that combining extracellular vesicles into bioactive multifunctional hydrogel should have great potential in achieving satisfactory locomotor recovery of central nervous system diseases. The novel FE hydrogel was designed for encapsulating the extracellular vesicles (FE@EVs). FE hydrogel exert the capabilities of temperature-responsive, injectable, adhesive and biocompatible. FE hydrogel with sustainable and long-term extracellular vesicle release for improving motor functional recovery after SCI. FE@EVs plays a vital role in pathological process of spinal cord injury in rats.
Collapse
|
89
|
Budgude P, Kale V, Vaidya A. Cryopreservation of mesenchymal stromal cell-derived extracellular vesicles using trehalose maintains their ability to expand hematopoietic stem cells in vitro. Cryobiology 2021; 98:152-163. [PMID: 33253747 DOI: 10.1016/j.cryobiol.2020.11.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/06/2020] [Accepted: 11/06/2020] [Indexed: 02/08/2023]
Abstract
The multitude of clinical trials using mesenchymal stromal cells (MSCs) has underscored their significance as a promising cell source for regenerative therapies. Most studies have however shown that MSCs get entrapped into the microvasculature of lungs, liver and spleen. In addition to intercellular communication, MSCs exert their effects in a paracrine manner by secretion of extracellular vesicles (EVs). The therapeutic effects of MSC-derived EVs have been examined in several diseases such as hepatic failure, liver injury, hematopoiesis etc. Therefore, optimization of cryopreservation strategies for the long-term storage of functional EVs could help in the development of off-the-shelf biologics. The aim of this study was to develop an optimal cryopreservation strategy for the efficient storage of both types of EVs - Microvesicles (MVs) and exosomes, independently, and to further examine the effect of the cryopreserved EVs on the ex vivo expansion of HSCs. MVs and exosomes were separately cryopreserved at different temperatures using PBS or PBS supplemented with trehalose (pTRE), and these cryopreserved EVs were then assessed for their functionality after revival. We found that addition of trehalose during cryopreservation helped in maintaining the morphology and functionality of the EVs, as assessed by their HSC-supportive potential, ability to expand phenotypically defined HSCs and ability to maintain the chemotactic migration potential of the HSCs co-cultured with them. This strategy could prove to be beneficial for facilitating the use of EVs as cell-free ready-to-use biologics for the ex vivo expansion of HSCs and in regenerative medicine.
Collapse
Affiliation(s)
- Pallavi Budgude
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India; Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, 412115, India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India
| | - Anuradha Vaidya
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India; Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, 412115, India.
| |
Collapse
|
90
|
Human amniotic membrane as a delivery vehicle for stem cell-based therapies. Life Sci 2021; 272:119157. [PMID: 33524418 DOI: 10.1016/j.lfs.2021.119157] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022]
Abstract
Stem cell-based therapy is known as a regenerative approach for a variety of diseases and tissue injuries. These cells exert their therapeutic effects through paracrine secretions namely extracellular vesicles. To achieve higher therapeutic potential, a variety of delivery routes have been tested in clinical and preclinical studies. Direct cell injection, intra-venous administration, and intra-arterial infusion are widely used methods of stem cells delivery but these methods are associated with several complications. As one of the most popular biological delivery systems, amniotic membrane has been widely utilized to support cell proliferation and differentiation therefore facilitating tissue regeneration without endangering the stem cells' viability. It is composed of several extracellular matrix components and growth factors. Due to these characteristics, amniotic membrane can mimic the stem cell's niche and can be an ideal carrier for stem cell transplantation. Here, we provide an overview of the recent progress, challenges, and future perspectives in the use of amniotic membrane as a delivery platform for stem cells.
Collapse
|
91
|
Liu Y, Zhuang X, Yu S, Yang N, Zeng J, Liu X, Chen X. Exosomes derived from stem cells from apical papilla promote craniofacial soft tissue regeneration by enhancing Cdc42-mediated vascularization. Stem Cell Res Ther 2021; 12:76. [PMID: 33482924 PMCID: PMC7821694 DOI: 10.1186/s13287-021-02151-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/07/2021] [Indexed: 12/18/2022] Open
Abstract
Background Reconstruction of complex critical-size defects (CSD) in the craniofacial region is a major challenge, and soft tissue regeneration is crucial in determining the therapeutic outcomes of craniofacial CSD. Stem cells from apical papilla (SCAP) are neural crest-derived mesenchymal stem cells (MSCs) that are homologous to cells in craniofacial tissue and represent a promising source for craniofacial tissue regeneration. Exosomes, which contain compound bioactive compounds, are the key factors in stem cell paracrine action. However, the roles of exosomes derived from SCAP (SCAP-Exo) in tissue regeneration are not fully understood. Here, we explored the effects and underlying mechanisms of SCAP-Exo on CSD in maxillofacial soft tissue. Methods SCAP-Exo were isolated and identified by transmission electron microscopy and nanoparticle tracking analysis. The effects of SCAP-Exo on wound healing and vascularization were detected by measuring the wound area and performing histological and immunofluorescence analysis on the palatal gingival CSD of mice. Real-time live-cell imaging and functional assays were used to assess the effects of SCAP-Exo on the biological functions of endothelial cells (ECs). Furthermore, the molecular mechanisms of SCAP-Exo-mediated EC angiogenesis in vitro were tested by immunofluorescence staining, Western blot, and pull-down assays. Finally, in vivo experiments were carried out to verify whether SCAP-Exo could affect vascularization and wound healing through cell division cycle 42 (Cdc42). Results We found that SCAP-Exo promoted tissue regeneration of palatal gingival CSD by enhancing vascularization in the early phase in vivo and that SCAP-Exo improved the angiogenic capacity of ECs in vitro. Mechanistically, SCAP-Exo elevated cell migration by improving cytoskeletal reorganization of ECs via Cdc42 signalling. Furthermore, we revealed that SCAP-Exo transferred Cdc42 into the cytoplasm of ECs and that the Cdc42 protein could be reused directly by recipient ECs, which resulted in the activation of Cdc42-dependent filopodium formation and elevation in cell migration of ECs. Conclusion This study demonstrated that SCAP-Exo had a superior effect on angiogenesis and effectively promoted craniofacial soft tissue regeneration. These data provide a new option for SCAP-Exo to be used in a cell-free approach to optimize tissue regeneration in the clinic. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02151-w.
Collapse
Affiliation(s)
- Yao Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, 117 Nanjing North Street, Shenyang, 110002, China.,Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xueying Zhuang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, 117 Nanjing North Street, Shenyang, 110002, China.,Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Si Yu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, 117 Nanjing North Street, Shenyang, 110002, China
| | - Ning Yang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, 117 Nanjing North Street, Shenyang, 110002, China
| | - Jianhong Zeng
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, 117 Nanjing North Street, Shenyang, 110002, China
| | - Xuemei Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, 117 Nanjing North Street, Shenyang, 110002, China.,Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xu Chen
- Department of Pediatric Dentistry, School and Hospital of Stomatology, China Medical University, 117 Nanjing North Street, Shenyang, 110002, China. .,Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China.
| |
Collapse
|
92
|
A versatile platform for generating engineered extracellular vesicles with defined therapeutic properties. Mol Ther 2021; 29:1729-1743. [PMID: 33484965 PMCID: PMC8116569 DOI: 10.1016/j.ymthe.2021.01.020] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/04/2020] [Accepted: 01/12/2021] [Indexed: 12/23/2022] Open
Abstract
Extracellular vesicles (EVs) are an important intercellular communication system facilitating the transfer of macromolecules between cells. Delivery of exogenous cargo tethered to the EV surface or packaged inside the lumen are key strategies for generating therapeutic EVs. We identified two “scaffold” proteins, PTGFRN and BASP1, that are preferentially sorted into EVs and enable high-density surface display and luminal loading of a wide range of molecules, including cytokines, antibody fragments, RNA binding proteins, vaccine antigens, Cas9, and members of the TNF superfamily. Molecules were loaded into EVs at high density and exhibited potent in vitro activity when fused to full-length or truncated forms of PTGFRN or BASP1. Furthermore, these engineered EVs retained pharmacodynamic activity in a variety of animal models. This engineering platform provides a simple approach to functionalize EVs with topologically diverse macromolecules and represents a significant advance toward unlocking the therapeutic potential of EVs.
Collapse
|
93
|
Ridzuan N, Zakaria N, Widera D, Sheard J, Morimoto M, Kiyokawa H, Mohd Isa SA, Chatar Singh GK, Then KY, Ooi GC, Yahaya BH. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles ameliorate airway inflammation in a rat model of chronic obstructive pulmonary disease (COPD). Stem Cell Res Ther 2021; 12:54. [PMID: 33436065 PMCID: PMC7805108 DOI: 10.1186/s13287-020-02088-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is an incurable and debilitating chronic disease characterized by progressive airflow limitation associated with abnormal levels of tissue inflammation. Therefore, stem cell-based approaches to tackle the condition are currently a focus of regenerative therapies for COPD. Extracellular vesicles (EVs) released by all cell types are crucially involved in paracrine, extracellular communication. Recent advances in the field suggest that stem cell-derived EVs possess a therapeutic potential which is comparable to the cells of their origin. METHODS In this study, we assessed the potential anti-inflammatory effects of human umbilical cord mesenchymal stem cell (hUC-MSC)-derived EVs in a rat model of COPD. EVs were isolated from hUC-MSCs and characterized by the transmission electron microscope, western blotting, and nanoparticle tracking analysis. As a model of COPD, male Sprague-Dawley rats were exposed to cigarette smoke for up to 12 weeks, followed by transplantation of hUC-MSCs or application of hUC-MSC-derived EVs. Lung tissue was subjected to histological analysis using haematoxylin and eosin staining, Alcian blue-periodic acid-Schiff (AB-PAS) staining, and immunofluorescence staining. Gene expression in the lung tissue was assessed using microarray analysis. Statistical analyses were performed using GraphPad Prism 7 version 7.0 (GraphPad Software, USA). Student's t test was used to compare between 2 groups. Comparison among more than 2 groups was done using one-way analysis of variance (ANOVA). Data presented as median ± standard deviation (SD). RESULTS Both transplantation of hUC-MSCs and application of EVs resulted in a reduction of peribronchial and perivascular inflammation, alveolar septal thickening associated with mononuclear inflammation, and a decreased number of goblet cells. Moreover, hUC-MSCs and EVs ameliorated the loss of alveolar septa in the emphysematous lung of COPD rats and reduced the levels of NF-κB subunit p65 in the tissue. Subsequent microarray analysis revealed that both hUC-MSCs and EVs significantly regulate multiple pathways known to be associated with COPD. CONCLUSIONS In conclusion, we show that hUC-MSC-derived EVs effectively ameliorate by COPD-induced inflammation. Thus, EVs could serve as a new cell-free-based therapy for the treatment of COPD.
Collapse
Affiliation(s)
- Noridzzaida Ridzuan
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200, Bertam, Penang, Malaysia
| | - Norashikin Zakaria
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200, Bertam, Penang, Malaysia
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine, School of Pharmacy, University of Reading, Reading, RG6 6AP, UK
| | - Jonathan Sheard
- Stem Cell Biology and Regenerative Medicine, School of Pharmacy, University of Reading, Reading, RG6 6AP, UK
| | - Mitsuru Morimoto
- RIKEN Centre for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuou-ku, Kobe, 650-0047, Japan
| | - Hirofumi Kiyokawa
- RIKEN Centre for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuou-ku, Kobe, 650-0047, Japan
| | - Seoparjoo Azmel Mohd Isa
- Department of Pathology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kubang Kerian, Malaysia
| | - Gurjeet Kaur Chatar Singh
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia
| | - Kong-Yong Then
- CryoCord Sdn Bhd, Bio-X Centre, 63000, Cyberjaya, Selangor, Malaysia
| | - Ghee-Chien Ooi
- CryoCord Sdn Bhd, Bio-X Centre, 63000, Cyberjaya, Selangor, Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200, Bertam, Penang, Malaysia.
- USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia.
| |
Collapse
|
94
|
Magarotto F, Sgrò A, Dorigo Hochuli AH, Andreetta M, Grassi M, Saggioro M, Nogara L, Tolomeo AM, Francescato R, Collino F, Germano G, Caicci F, Maghin E, Piccoli M, Jurga M, Blaauw B, Gamba P, Muraca M, Pozzobon M. Muscle functional recovery is driven by extracellular vesicles combined with muscle extracellular matrix in a volumetric muscle loss murine model. Biomaterials 2021; 269:120653. [PMID: 33461058 DOI: 10.1016/j.biomaterials.2021.120653] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/26/2020] [Accepted: 01/02/2021] [Indexed: 12/23/2022]
Abstract
Biological scaffolds derived from decellularized tissues are being investigated as a promising approach to repair volumetric muscle losses (VML). Indeed, extracellular matrix (ECM) from decellularized tissues is highly biocompatible and mimics the original tissue. However, the development of fibrosis and the muscle stiffness still represents a major problem. Intercellular signals mediating tissue repair are conveyed via extracellular vesicles (EVs), biologically active nanoparticles secreted by the cells. This work aimed at using muscle ECM and human EVs derived from Wharton Jelly mesenchymal stromal cells (MSC EVs) to boost tissue regeneration in a VML murine model. Mice transplanted with muscle ECM and treated with PBS or MSC EVs were analyzed after 7 and 30 days. Flow cytometry, tissue analysis, qRT-PCR and physiology test were performed. We demonstrated that angiogenesis and myogenesis were enhanced while fibrosis was reduced after EV treatment. Moreover, the inflammation was directed toward tissue repair. M2-like, pro-regenerative macrophages were significantly increased in the MSC EVs treated group compared to control. Strikingly, the histological improvements were associated with enhanced functional recovery. These results suggest that human MSC EVs can be a naturally-derived boost able to ameliorate the efficacy of tissue-specific ECM in muscle regeneration up to the restored tissue function.
Collapse
Affiliation(s)
- Fabio Magarotto
- Stem Cells and Regenerative Medicine Lab, Institute of Pediatric Research Città Della Speranza, Padova, Italy; Department of Women and Children Health, University of Padova, Italy
| | - Alberto Sgrò
- Department of Women and Children Health, University of Padova, Italy
| | | | - Marina Andreetta
- Department of Women and Children Health, University of Padova, Italy
| | - Michele Grassi
- Department of Women and Children Health, University of Padova, Italy
| | - Mattia Saggioro
- Stem Cells and Regenerative Medicine Lab, Institute of Pediatric Research Città Della Speranza, Padova, Italy; Department of Women and Children Health, University of Padova, Italy
| | - Leonardo Nogara
- Biomedical Sciences Department, University of Padova, Italy; Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Anna Maria Tolomeo
- Department of Women and Children Health, University of Padova, Italy; L.i.f.e.L.a.b. Program, Consorzio per La Ricerca Sanitaria (CORIS), Veneto Region, Padova, Italy
| | - Riccardo Francescato
- Stem Cells and Regenerative Medicine Lab, Institute of Pediatric Research Città Della Speranza, Padova, Italy
| | - Federica Collino
- Laboratory of Translational Research in Paediatric Nephro-urology, Fondazione Ca' Granada IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - Giuseppe Germano
- Institute of Pediatric Research Città Della Speranza, Padova, Italy
| | | | - Edoardo Maghin
- Department of Women and Children Health, University of Padova, Italy; Tissue Engineering Lab, Institute of Pediatric Research Città Della Speranza, Padova, Italy
| | - Martina Piccoli
- Tissue Engineering Lab, Institute of Pediatric Research Città Della Speranza, Padova, Italy
| | | | - Bert Blaauw
- Biomedical Sciences Department, University of Padova, Italy; Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Piergiorgio Gamba
- Department of Women and Children Health, University of Padova, Italy
| | - Maurizio Muraca
- Department of Women and Children Health, University of Padova, Italy; Institute of Pediatric Research Città Della Speranza, Padova, Italy; L.i.f.e.L.a.b. Program, Consorzio per La Ricerca Sanitaria (CORIS), Veneto Region, Padova, Italy
| | - Michela Pozzobon
- Stem Cells and Regenerative Medicine Lab, Institute of Pediatric Research Città Della Speranza, Padova, Italy; Department of Women and Children Health, University of Padova, Italy.
| |
Collapse
|
95
|
Qin B, Zhang Q, Chen D, Yu HY, Luo AX, Suo LP, Cai Y, Cai DY, Luo J, Huang JF, Xiong K. Extracellular vesicles derived from mesenchymal stem cells: A platform that can be engineered. Histol Histopathol 2021; 36:615-632. [PMID: 33398872 DOI: 10.14670/hh-18-297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells play an important role in tissue damage and repair. This role is mainly due to a paracrine mechanism, and extracellular vesicles (EVs) are an important part of the paracrine function. EVs play a vital role in many aspects of cell homeostasis, physiology, and pathology, and EVs can be used as clinical biomarkers, vaccines, or drug delivery vehicles. A large number of studies have shown that EVs derived from mesenchymal stem cells (MSC-EVs) play an important role in the treatment of various diseases. However, the problems of low production, low retention rate, and poor targeting of MSC-EVs are obstacles to current clinical applications. The engineering transformation of MSC-EVs can make up for those shortcomings, thereby improving treatment efficiency. This review summarizes the latest research progress of MSC-EV direct and indirect engineering transformation from the aspects of improving MSC-EV retention rate, yield, targeting, and MSC-EV visualization research, and proposes some feasible MSC-EV engineering methods of transformation.
Collapse
Affiliation(s)
- Bo Qin
- Hubei Polytechnic University School of Medicine, Huangshi, Hubei, China
| | - Qi Zhang
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Dan Chen
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Hai-Yang Yu
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Ai-Xiang Luo
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Liang-Peng Suo
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Yan Cai
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - De-Yang Cai
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jia Luo
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Ju-Fang Huang
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, China.
| | - Kun Xiong
- Department of Neurobiology and Human Anatomy, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Ophthalmology, Changsha, Hunan, China.
| |
Collapse
|
96
|
Küçükgüven MB, Çelebi-Saltik B. Stem Cell Based Exosomes: Are They Effective in Disease or Health? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1341:45-65. [PMID: 33782904 DOI: 10.1007/5584_2021_630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Exosomes are nano-sized vesicles involved in intercellular communication via delivery of molecules including lipids, nucleic acids, proteins, or other cellular components to distant or neighboring sites. Their ability to pass biological barriers, stability in physiological fluids without degradation, and distinctive affinity to target cells make exosomes very remarkable therapeutic vehicles. Virus-based approaches are some of the most widely used gene therapy methods; however, there are many issues need to be clarified such as high immunogenicity. Using of the exosomes procures the functional transfer of their cargo with minimal intervention from the immune system and it has been reported to be secure and well-tolerated. When the regenerative medicine is taken into consideration, stem cell-based approaches have been aimed to utilize but the general efficacy and safety profile of stem cell therapy has still not been enlightened. At this point, stem cell-derived exosomes exhibit a way to procure cell-free regenerative medicine with their unique characteristics. Exosomes are considered as appropriate and highly stable biological nano-vectors taking part in a wide variety of healthy and pathological processes for advanced targeted therapies. However, there are still crucial obstacles to achieve efficient isolation of large amount of specific and pure exosomes. Thus, large-scale exosome production under good manufacturing practice is required. The purpose of this review is to focus on stem cell-based exosomes for gene delivery and to introduce synthetic exosome-mimics as a potential alternative in the field of targeted gene therapies. Further, we aim to highlight the biobanking and large-scale manufacturing methods of exosomes.
Collapse
Affiliation(s)
- Meriç Bilgiç Küçükgüven
- Department of Oral and Maxillofacial Surgery, Hacettepe University Faculty of Dentistry, Ankara, Turkey.,Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey.,Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| | - Betül Çelebi-Saltik
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey. .,Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
97
|
Alwjwaj M, Kadir RRA, Bayraktutan U. The secretome of endothelial progenitor cells: a potential therapeutic strategy for ischemic stroke. Neural Regen Res 2021; 16:1483-1489. [PMID: 33433461 PMCID: PMC8323700 DOI: 10.4103/1673-5374.303012] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ischemic stroke continues to be a leading cause of mortality and morbidity in the world. Despite recent advances in the field of stroke medicine, thrombolysis with recombinant tissue plasminogen activator remains as the only pharmacological therapy for stroke patients. However, due to short therapeutic window (4.5 hours of stroke onset) and increased risk of hemorrhage beyond this point, each year globally less than 1% of stroke patients receive this therapy which necessitate the discovery of safe and efficacious therapeutics that can be used beyond the acute phase of stroke. Accumulating evidence indicates that endothelial progenitor cells (EPCs), equipped with an inherent capacity to migrate, proliferate and differentiate, may be one such therapeutics. However, the limited availability of EPCs in peripheral blood and early senescence of few isolated cells in culture conditions adversely affect their application as effective therapeutics. Given that much of the EPC-mediated reparative effects on neurovasculature is realized by a wide range of biologically active substances released by these cells, it is possible that EPC-secretome may serve as an important therapeutic after an ischemic stroke. In light of this assumption, this review paper firstly discusses the main constituents of EPC-secretome that may exert the beneficial effects of EPCs on neurovasculature, and then reviews the currently scant literature that focuses on its therapeutic capacity.
Collapse
Affiliation(s)
- Mansour Alwjwaj
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Rais Reskiawan A Kadir
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Ulvi Bayraktutan
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| |
Collapse
|
98
|
Anwar I, Ashfaq UA, Shokat Z. Therapeutic Potential of Umbilical Cord Stem Cells for Liver Regeneration. Curr Stem Cell Res Ther 2020; 15:219-232. [PMID: 32077830 DOI: 10.2174/1568026620666200220122536] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/16/2019] [Accepted: 08/08/2019] [Indexed: 01/18/2023]
Abstract
The liver is a vital organ for life and the only internal organ that is capable of natural regeneration. Although the liver has high regeneration capacity, excessive hepatocyte death can lead to liver failure. Various factors can lead to liver damage including drug abuse, some natural products, alcohol, hepatitis, and autoimmunity. Some models for studying liver injury are APAP-based model, Fas ligand (FasL), D-galactosamine/endotoxin (Gal/ET), Concanavalin A, and carbon tetrachloride-based models. The regeneration of the liver can be carried out using umbilical cord blood stem cells which have various advantages over other stem cell types used in liver transplantation. UCB-derived stem cells lack tumorigenicity, have karyotype stability and high immunomodulatory, low risk of graft versus host disease (GVHD), low risk of transmitting somatic mutations or viral infections, and low immunogenicity. They are readily available and their collection is safe and painless. This review focuses on recent development and modern trends in the use of umbilical cord stem cells for the regeneration of liver fibrosis.
Collapse
Affiliation(s)
- Ifrah Anwar
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Usman A Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Zeeshan Shokat
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| |
Collapse
|
99
|
De Luna A, Otahal A, Nehrer S. Mesenchymal Stromal Cell-Derived Extracellular Vesicles - Silver Linings for Cartilage Regeneration? Front Cell Dev Biol 2020; 8:593386. [PMID: 33363147 PMCID: PMC7758223 DOI: 10.3389/fcell.2020.593386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/19/2020] [Indexed: 01/15/2023] Open
Abstract
As the world's population is aging, the incidence of the degenerative disease Osteoarthritis (OA) is increasing. Current treatment options of OA focus on the alleviation of the symptoms including pain and inflammation rather than on restoration of the articular cartilage. Cell-based therapies including the application of mesenchymal stromal cells (MSCs) have been a promising tool for cartilage regeneration approaches. Due to their immunomodulatory properties, their differentiation potential into cells of the mesodermal lineage as well as the plurality of sources from which they can be isolated, MSCs have been applied in a vast number of studies focusing on the establishment of new treatment options for Osteoarthritis. Despite promising outcomes in vitro and in vivo, applications of MSCs are connected with teratoma formation, limited lifespan of differentiated cells as well as rejection of the cells after transplantation, highlighting the need for new cell free approaches harboring the beneficial properties of MSCs. It has been demonstrated that the regenerative potential of MSCs is mediated by the release of paracrine factors rather than by differentiation into cells of the desired tissue. Besides soluble factors, extracellular vesicles are the major component of a cell's secretome. They represent novel mechanisms by which (pathogenic) signals can be communicated between cell types as they deliver bioactive molecules (nucleic acids, proteins, lipids) from the cell of origin to the target cell leading to specific biological processes upon uptake. This review will give an overview about extracellular vesicles including general characteristics, isolation methods and characterization approaches. Furthermore, the role of MSC-derived extracellular vesicles in in vitro and in vivo studies for cartilage regeneration will be summarized with special focus on transported miRNA which either favored the progression of OA or protected the cartilage from degradation. In addition, studies will be reviewed investigating the impact of MSC-derived extracellular vesicles on inflammatory arthritis. As extracellular vesicles are present in all body fluids, their application as potential biomarkers for OA will also be discussed in this review. Finally, studies exploring the combination of MSC-derived extracellular vesicles with biomaterials for tissue engineering approaches are summarized.
Collapse
Affiliation(s)
- Andrea De Luna
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| | | | | |
Collapse
|
100
|
Oesterreicher J, Pultar M, Schneider J, Mühleder S, Zipperle J, Grillari J, Holnthoner W. Fluorescence-Based Nanoparticle Tracking Analysis and Flow Cytometry for Characterization of Endothelial Extracellular Vesicle Release. Int J Mol Sci 2020; 21:E9278. [PMID: 33291792 PMCID: PMC7731108 DOI: 10.3390/ijms21239278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022] Open
Abstract
As extracellular vesicles (EVs) have become a prominent topic in life sciences, a growing number of studies are published on a regular basis addressing their biological relevance and possible applications. Nevertheless, the fundamental question of the true vesicular nature as well as possible influences on the EV secretion behavior have often been not adequately addressed. Furthermore, research regarding endothelial cell-derived EVs (EndoEVs) often focused on the large vesicular fractions comprising of microvesicles (MV) and apoptotic bodies. In this study we aimed to further extend the current knowledge of the influence of pre-isolation conditions, such as cell density and conditioning time, on EndoEV release from human umbilical vein endothelial cells (HUVECs). We combined fluorescence nanoparticle tracking analysis (NTA) and the established fluorescence-triggered flow cytometry (FT-FC) protocol to allow vesicle-specific detection and characterization of size and surface markers. We found significant effects of cell density and conditioning time on both abundance and size distribution of EndoEVs. Additionally, we present detailed information regarding the surface marker display on EVs from different fractions and size ranges. Our data provide crucial relevance for future projects aiming to elucidate EV secretion behavior of endothelial cells. Moreover, we show that the influence of different conditioning parameters on the nature of EndoEVs has to be considered.
Collapse
Affiliation(s)
- Johannes Oesterreicher
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Centre, 1200 Vienna, Austria; (J.O.); (M.P.); (J.S.); (J.Z.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Marianne Pultar
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Centre, 1200 Vienna, Austria; (J.O.); (M.P.); (J.S.); (J.Z.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Jaana Schneider
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Centre, 1200 Vienna, Austria; (J.O.); (M.P.); (J.S.); (J.Z.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Severin Mühleder
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain;
| | - Johannes Zipperle
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Centre, 1200 Vienna, Austria; (J.O.); (M.P.); (J.S.); (J.Z.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Centre, 1200 Vienna, Austria; (J.O.); (M.P.); (J.S.); (J.Z.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, 1090 Vienna, Austria
| | - Wolfgang Holnthoner
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Centre, 1200 Vienna, Austria; (J.O.); (M.P.); (J.S.); (J.Z.); (J.G.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|